1
|
Priya, Kumar A, Kumar D. Molecular heterogeneity and MYC dysregulation in triple-negative breast cancer: genomic advances and therapeutic implications. 3 Biotech 2025; 15:33. [PMID: 39777154 PMCID: PMC11700964 DOI: 10.1007/s13205-024-04195-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a diverse range of molecular features that have been extensively studied. MYC plays a critical role in regulating metabolism, differentiation, proliferation, cell growth, and apoptosis. Dysregulation of MYC is associated with poor prognosis and contributes to the development and progression of breast cancer. A particularly intriguing aspect of TNBC is its association with tumors in BRCA1 mutation carriers, especially in younger women. MYC may also contribute to resistance to adjuvant treatments. For TNBC, targeting MYC-regulated pathways in combination with inhibitors of other carcinogenic pathways offers a promising therapeutic approach. Several signaling pathways regulate TNBC, and targeting these pathways could lead to effective therapeutic strategies for breast cancer. Advances in genomic tools, such as CRISPR-Cas9, next-generation sequencing, and whole-exome sequencing, are revolutionizing breast cancer diagnoses. These technologies have significantly enhanced our understanding of MYC oncogenesis, particularly through CRISPR-Cas9 and NGS. Targeting MYC and its partner MAX could provide valuable insights into TNBC. Moreover, the therapeutic potential of targeting MYC-driven signaling mechanisms and their interactions with other oncogenic pathways, including PI3K/AKT/mTOR and Wnt/β-catenin, is increasingly recognized. Next-generation sequencing and CRISPR-Cas9 represent significant breakthroughs in genomic tools that open new opportunities to explore MYC's role in TNBC and facilitate the development of personalized treatment plans. This review discusses the future clinical applications of personalized treatment strategies for patients with TNBC.
Collapse
Affiliation(s)
- Priya
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| | - Arun Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar 801505 India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| |
Collapse
|
2
|
Mai N, Abuhadra N, Jhaveri K. Molecularly Targeted Therapies for Triple Negative Breast Cancer: History, Advances, and Future Directions. Clin Breast Cancer 2023; 23:784-799. [PMID: 37336650 DOI: 10.1016/j.clbc.2023.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 06/21/2023]
Abstract
Triple negative breast cancer (TNBC) remains the subtype with poorest prognosis. Despite the subtype's heterogeneity, there is still a paucity in effective targeted therapeutics that offer both good efficacy and tolerability, and chemotherapy remains the backbone of modern TNBC therapy. In the past few years, immunotherapy as well as novel therapeutic modalities like antibody-drug conjugates (ADCs) have shown clinical benefit and have been FDA approved in various clinical stages of unselected TNBC. However, there has not been similar advancement in molecularly targeted therapies, especially when compared to advancements seen in hormone receptor (HR)-positive or HER2-positive breast cancer. PARP inhibitors have been approved for BRCA-mutated TNBC, but responses are short-lived, and resistance remains a barrier for current treatment. PI3K pathway inhibitors approved in HR+ breast cancer has not worked for TNBC and continue to have significant dose-limiting adverse effects. EGFR inhibition has been thoroughly explored in TNBC, but all trials so far have shown minimal efficacy. Nevertheless, despite these setbacks, current research in targeted therapy for TNBC holds great promise in overcoming the barriers of the past and developing novel therapeutic approaches for the future. In this review, we describe molecular targets both identified and validated in the treatment of TNBC, discuss the historical efforts towards development of targeted agents and current areas of improvement, and address promising advances that have the potential to improve outcomes in this heterogenous and aggressive breast cancer subtype. Immunotherapy, ADCs, and AR targeting will be discussed in separate reviews of this edition.
Collapse
Affiliation(s)
- Nicholas Mai
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nour Abuhadra
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
3
|
Singh H. Role of Molecular Targeted Therapeutic Drugs in Treatment of Breast Cancer: A Review Article. Glob Med Genet 2023; 10:79-86. [PMID: 37228871 PMCID: PMC10205396 DOI: 10.1055/s-0043-57247] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023] Open
Abstract
Breast cancer is a multifactor, multistage, and heterogeneous disease. Systemic treatment of breast cancer has changed significantly over the last decade. With a better knowledge of the pathogenesis, researchers and scientists have discovered numerous signaling pathways and synonymous therapeutic targets in breast cancer. Because of the molecular nature of breast cancer, which makes it difficult to understand, previous attempts to treat or prevent it have failed. However, recent decades have provided effective therapeutic targets for treatment. In this review, literature or information on various targeted therapy for breast cancer is discussed. English language articles were explored in numerous directory or databases like PubMed, Web of Sciences, Google Scholar, ScienceDirect, and Scopus. The important keywords used for searching databases are "Breast cancer," "Targeted therapy in breast cancer," "Therapeutic drugs in breast cancer," and "Molecular targets in breast cancer."
Collapse
Affiliation(s)
- Himanshu Singh
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Index Institute of Dental Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
4
|
Drago JZ, Ferraro E, Abuhadra N, Modi S. Beyond HER2: Targeting the ErbB receptor family in breast cancer. Cancer Treat Rev 2022; 109:102436. [PMID: 35870237 PMCID: PMC10478787 DOI: 10.1016/j.ctrv.2022.102436] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/07/2022] [Accepted: 07/10/2022] [Indexed: 11/28/2022]
Abstract
Targeting the HER2 oncogene represents one of the greatest advances in the treatment of breast cancer. HER2 is one member of the ERBB-receptor family, which includes EGFR (HER1), HER3 and HER4. In the presence or absence of underling genomic aberrations such as mutations or amplification events, intricate interactions between these proteins on the cell membrane lead to downstream signaling that encourages cancer growth and proliferation. In this Review, we contextualize efforts to pharmacologically target the ErbB receptor family beyond HER2, with a focus on EGFR and HER3. Preclinical and clinical efforts are synthesized. We discuss successes and failures of this approach to date, summarize lessons learned, and propose a way forward that invokes new therapeutic modalities such as antibody drug conjugates (ADCs), combination strategies, and patient selection through rational biomarkers.
Collapse
Affiliation(s)
- Joshua Z Drago
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA.
| | - Emanuela Ferraro
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nour Abuhadra
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weil Cornell Medicine, New York, NY, USA
| |
Collapse
|
5
|
Radovich M, Solzak JP, Wang CJ, Hancock BA, Badve S, Althouse SK, Bray SM, Storniolo AMV, Ballinger TJ, Schneider BP, Miller KD. Initial Phase I Safety Study of Gedatolisib plus Cofetuzumab Pelidotin for Patients with Metastatic Triple-Negative Breast Cancer. Clin Cancer Res 2022; 28:3235-3241. [PMID: 35551360 PMCID: PMC9357180 DOI: 10.1158/1078-0432.ccr-21-3078] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/18/2021] [Accepted: 05/10/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE The PI3K pathway is dysregulated in the majority of triple-negative breast cancers (TNBC), yet single-agent inhibition of PI3K has been ineffective in TNBC. PI3K inhibition leads to an immediate compensatory upregulation of the Wnt pathway. Dual targeting of both pathways is highly synergistic against TNBC models in vitro and in vivo. We initiated a phase I clinical trial combining gedatolisib, a pan-class I isoform PI3K/mTOR inhibitor, and cofetuzumab pelidotin, an antibody-drug conjugate against the cell-surface PTK7 protein (Wnt pathway coreceptor) with an auristatin payload. PATIENTS AND METHODS Participants (pt) had metastatic TNBC or estrogen receptor (ER) low (ER and PgR < 5%, HER2-negative) breast cancer, and had received at least one prior chemotherapy for advanced disease. The primary objective was safety. Secondary endpoints included overall response rate (ORR), clinical benefit at 18 weeks (CB18), progression-free survival (PFS), and correlative analyses. RESULTS A total of 18 pts were enrolled in three dose cohorts: gedatolisib 110 mg weekly + cofetuzumab pelidotin 1.4 mg/kg every 3 weeks (n = 4), 180 mg + 1.4 mg/kg (n = 3), and 180 mg + 2.8 mg/kg (n = 11). Nausea, anorexia, fatigue, and mucositis were common but rarely reached ≥grade 3 severity. Myelosuppression was uncommon. ORR was 16.7% (3/18). An additional 3 pts had stable disease (of these 2 had stable disease for >18 weeks); CB18 was 27.8%. Median PFS was 2.0 months (95% confidence interval for PFS: 1.2-6.2). Pts with clinical benefit were enriched with genomic alterations in the PI3K and PTK7 pathways. CONCLUSIONS The combination of gedatolisib + cofetuzumab pelidotin was well tolerated and demonstrated promising clinical activity. Further investigation of this drug combination in metastatic TNBC is warranted.
Collapse
Affiliation(s)
- Milan Radovich
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Surgery, Division of Surgical Oncology, Indiana University School of Medicine
| | - Jeffrey P. Solzak
- Department of Surgery, Division of Surgical Oncology, Indiana University School of Medicine
| | - Chao J. Wang
- Department of Surgery, Division of Surgical Oncology, Indiana University School of Medicine
| | - Bradley A. Hancock
- Department of Surgery, Division of Surgical Oncology, Indiana University School of Medicine
| | - Sunil Badve
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Pathology, Indiana University School of Medicine
| | - Sandra K. Althouse
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Biostatistics and Data Health Science, Indiana University School of Medicine
| | | | - Anna Maria V. Storniolo
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine
| | - Tarah J. Ballinger
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine
| | - Bryan P. Schneider
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine
| | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
- Department of Medicine, Division of Hematology/Oncology, Indiana University School of Medicine
| |
Collapse
|
6
|
de Araújo RA, da Luz FAC, da Costa Marinho E, Nascimento CP, de Andrade Marques L, Delfino PFR, Antonioli RM, Araújo BJ, da Silva ACAL, Dos Reis Monteiro MLG, Neto MB, Silva MJB. Epidermal growth factor receptor (EGFR) expression in the serum of patients with triple-negative breast carcinoma: prognostic value of this biomarker. Ecancermedicalscience 2022; 16:1431. [PMID: 36158981 PMCID: PMC9458269 DOI: 10.3332/ecancer.2022.1431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background Epidermal growth factor receptor (EGFR) overexpression has been considered a poor prognostic factor in breast cancer. Methodology A prospective study of 206 women with breast cancer analysed by stages (I, II, III and IV) and by immunohistochemical subtype (Luminal A, Luminal B, HER2+ and triple-negative (TN)); 89 healthy controls with normal recent mammography were included. The EGFR measured in the serum (sEGFR) was detected by the Enzyme-Linked Immunosorbent Assay (ELISA) method (R&D Systems kit DY231) collected by blood before any treatment in patients. Kaplan-Meier method and Cox regression were carried out to obtain the prognostic value, considering significance if p < 0.05. Results With a median follow-up of 36.6 months, 47 deaths occurred. Multivariable Cox regression showed difference of overall survival (OS) associated with sEGFR levels (sEGFR ≤ or > 47.8 ng/mL) in patients with TN cancers, but not of Luminal A, Luminal B or HER2+ subtypes; adjusted by stage, the death risk increased by approximately 415% [hazard ratio (HR): 5.149 (1.900-13.955), p = 0.001] for patients with sEGFR > 47.8 ng/mL compared to patients with a lower sEGFR value. There was no significant correlation of sEGFR with staging, histological tumour grade (G1/G2/G3), Ki67 (< or ≥14%) or body mass index. Conclusions Increased sEGFR expression in patients with TN tumours is a significant predictor of lower OS and its quantification is inexpensive and straightforward.
Collapse
Affiliation(s)
- Rogério Agenor de Araújo
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4653-6786
| | - Felipe Andrés Cordero da Luz
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-9381-4913
| | - Eduarda da Costa Marinho
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-1307-9104
| | - Camila Piqui Nascimento
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-0955-8559
| | - Lara de Andrade Marques
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2734-8352
| | - Patrícia Ferreira Ribeiro Delfino
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0002-2196-9318
| | - Rafael Mathias Antonioli
- Cancer Research and Prevention Nucleus, Grupo Luta Pela Vida, Cancer Hospital in Uberlândia, Uberlândia, MG, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-3886-1562
| | - Breno Jeha Araújo
- São Paulo State Cancer Institute of the Medical School of the University of São Paulo, São Paulo, SP, CEP 38405-302, Brazil
- https://orcid.org/0000-0003-4892-9911
| | - Ana Cristina Araújo Lemos da Silva
- Federal University of Uberlândia, Avenida Pará, Bloco 2U, 1720, Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil
- https://orcid.org/0000-0002-8220-938X
| | | | - Morun Bernardino Neto
- Department of Basic and Environmental Sciences, University of São Paulo, Lorena, SP, CEP 12602-810, Brazil
- https://orcid.org/0000-0003-4292-7800
| | - Marcelo José Barbosa Silva
- Laboratory of Tumor Biomarkers and Osteoimmunology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, MG, CEP 38405-320, Brazil
- https://orcid.org/0000-0002-5807-4286
| |
Collapse
|
7
|
Chowdhury P, Ghosh U, Samanta K, Jaggi M, Chauhan SC, Yallapu MM. Bioactive nanotherapeutic trends to combat triple negative breast cancer. Bioact Mater 2021; 6:3269-3287. [PMID: 33778204 PMCID: PMC7970221 DOI: 10.1016/j.bioactmat.2021.02.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 02/28/2021] [Indexed: 02/09/2023] Open
Abstract
The management of aggressive breast cancer, particularly, triple negative breast cancer (TNBC) remains a formidable challenge, despite treatment advancement. Although newer therapies such as atezolizumab, olaparib, and sacituzumab can tackle the breast cancer prognosis and/or progression, but achieved limited survival benefit(s). The current research efforts are aimed to develop and implement strategies for improved bioavailability, targetability, reduce systemic toxicity, and enhance therapeutic outcome of FDA-approved treatment regimen. This review presents various nanoparticle technology mediated delivery of chemotherapeutic agent(s) for breast cancer treatment. This article also documents novel strategies to employ cellular and cell membrane cloaked (biomimetic) nanoparticles for effective clinical translation. These technologies offer a safe and active targeting nanomedicine for effective management of breast cancer, especially TNBC.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Upasana Ghosh
- Department of Biomedical Engineering, School of Engineering, Rutgers University, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Kamalika Samanta
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C. Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
8
|
You KS, Yi YW, Cho J, Park JS, Seong YS. Potentiating Therapeutic Effects of Epidermal Growth Factor Receptor Inhibition in Triple-Negative Breast Cancer. Pharmaceuticals (Basel) 2021; 14:589. [PMID: 34207383 PMCID: PMC8233743 DOI: 10.3390/ph14060589] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a subset of breast cancer with aggressive characteristics and few therapeutic options. The lack of an appropriate therapeutic target is a challenging issue in treating TNBC. Although a high level expression of epidermal growth factor receptor (EGFR) has been associated with a poor prognosis among patients with TNBC, targeted anti-EGFR therapies have demonstrated limited efficacy for TNBC treatment in both clinical and preclinical settings. However, with the advantage of a number of clinically approved EGFR inhibitors (EGFRis), combination strategies have been explored as a promising approach to overcome the intrinsic resistance of TNBC to EGFRis. In this review, we analyzed the literature on the combination of EGFRis with other molecularly targeted therapeutics or conventional chemotherapeutics to understand the current knowledge and to provide potential therapeutic options for TNBC treatment.
Collapse
Affiliation(s)
- Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
| | - Yong Weon Yi
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeonghee Cho
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea;
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 3116, Chungcheongnam-do, Korea
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (J.C.)
| |
Collapse
|
9
|
Škubník J, Pavlíčková V, Ruml T, Rimpelová S. Current Perspectives on Taxanes: Focus on Their Bioactivity, Delivery and Combination Therapy. PLANTS (BASEL, SWITZERLAND) 2021; 10:569. [PMID: 33802861 PMCID: PMC8002726 DOI: 10.3390/plants10030569] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/09/2021] [Accepted: 03/15/2021] [Indexed: 12/14/2022]
Abstract
Taxanes, mainly paclitaxel and docetaxel, the microtubule stabilizers, have been well known for being the first-line therapy for breast cancer for more than the last thirty years. Moreover, they have been also used for the treatment of ovarian, hormone-refractory prostate, head and neck, and non-small cell lung carcinomas. Even though paclitaxel and docetaxel significantly enhance the overall survival rate of cancer patients, there are some limitations of their use, such as very poor water solubility and the occurrence of severe side effects. However, this is what pushes the research on these microtubule-stabilizing agents further and yields novel taxane derivatives with significantly improved properties. Therefore, this review article brings recent advances reported in taxane research mainly in the last two years. We focused especially on recent methods of taxane isolation, their mechanism of action, development of their novel derivatives, formulations, and improved tumor-targeted drug delivery. Since cancer cell chemoresistance can be an unsurpassable hurdle in taxane administration, a significant part of this review article has been also devoted to combination therapy of taxanes in cancer treatment. Last but not least, we summarize ongoing clinical trials on these compounds and bring a perspective of advancements in this field.
Collapse
Affiliation(s)
| | | | | | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 3, 166 28 Prague 6, Czech Republic; (J.Š.); (V.P.); (T.R.)
| |
Collapse
|
10
|
Maennling AE, Tur MK, Niebert M, Klockenbring T, Zeppernick F, Gattenlöhner S, Meinhold-Heerlein I, Hussain AF. Molecular Targeting Therapy against EGFR Family in Breast Cancer: Progress and Future Potentials. Cancers (Basel) 2019; 11:cancers11121826. [PMID: 31756933 PMCID: PMC6966464 DOI: 10.3390/cancers11121826] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) family contains four transmembrane tyrosine kinases (EGFR1/ErbB1, Her2/ErbB2, Her3/ErbB3 and Her4/ErbB4) and 13 secreted polypeptide ligands. EGFRs are overexpressed in many solid tumors, including breast, pancreas, head-and-neck, prostate, ovarian, renal, colon, and non-small-cell lung cancer. Such overexpression produces strong stimulation of downstream signaling pathways, which induce cell growth, cell differentiation, cell cycle progression, angiogenesis, cell motility and blocking of apoptosis.The high expression and/or functional activation of EGFRs correlates with the pathogenesis and progression of several cancers, which make them attractive targets for both diagnosis and therapy. Several approaches have been developed to target these receptors and/or the EGFR modulated effects in cancer cells. Most approaches include the development of anti-EGFRs antibodies and/or small-molecule EGFR inhibitors. This review presents the state-of-the-art and future prospects of targeting EGFRs to treat breast cancer.
Collapse
Affiliation(s)
- Amaia Eleonora Maennling
- Department of Gynecology and Obstetrics, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Mehmet Kemal Tur
- Institute of Pathology, University Hospital Giessen, Justus-Liebig-University Giessen, Langhanssstr. 10, 35392 Giessen, Germany
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine and Life Science, Maastricht University, Universiteitssingel 40, 6229 MD Maastricht, The Netherlands
| | - Marcus Niebert
- Department of Molecular Cytology and Functional Genomics, Institute of Pathology, University Hospital Giessen, Justus-Liebig-University Giessen, Langhanssstr. 10, 35392 Giessen, Germany
| | - Torsten Klockenbring
- Department of Biological Sensing and Detection, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Forckenbeckstrasse 6, 52074 Aachen, Germany
| | - Felix Zeppernick
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Stefan Gattenlöhner
- Institute of Pathology, University Hospital Giessen, Justus-Liebig-University Giessen, Langhanssstr. 10, 35392 Giessen, Germany
| | - Ivo Meinhold-Heerlein
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Ahmad Fawzi Hussain
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
- Correspondence: ; Tel.: +49-64199930570
| |
Collapse
|
11
|
Al-Mahmood S, Sapiezynski J, Garbuzenko OB, Minko T. Metastatic and triple-negative breast cancer: challenges and treatment options. Drug Deliv Transl Res 2018; 8:1483-1507. [PMID: 29978332 PMCID: PMC6133085 DOI: 10.1007/s13346-018-0551-3] [Citation(s) in RCA: 343] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The major current conventional types of metastatic breast cancer (MBC) treatments include surgery, radiation, hormonal therapy, chemotherapy, or immunotherapy. Introducing biological drugs, targeted treatment and gene therapy can potentially reduce the mortality and improve the quality of life in patients with MBC. However, combination of several types of treatment is usually recommended. Triple negative breast cancer (TNBC) accounts for 10-20% of all cases of breast carcinoma and is characterized by the low expression of progesterone receptor (PR), estrogen receptor (ER), and human epidermal growth factor receptor 2 (HER2). Consequently, convenient treatments used for MBC that target these receptors are not effective for TNBC which therefore requires special treatment approaches. This review discusses the occurrence of MBC, the prognosis and predictive biomarkers of MBC, and focuses on the novel advanced tactics for treatment of MBC and TNBC. Nanotechnology-based combinatorial approach for the suppression of EGFR by siRNA and gifitinib is described.
Collapse
Affiliation(s)
- Sumayah Al-Mahmood
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854-8020, USA
| | - Justin Sapiezynski
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854-8020, USA
| | - Olga B Garbuzenko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854-8020, USA
| | - Tamara Minko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ, 08854-8020, USA.
- Rutgers Cancer Institute, New Brunswick, NJ, 08903, USA.
- Environmental and Occupational Health Sciences Institute, Rutgers, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
12
|
Therapeutic Antibodies in Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 917:95-120. [PMID: 27236554 DOI: 10.1007/978-3-319-32805-8_6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The therapeutic arsenal in solid tumors comprises different anticancer strategies with diverse chemotherapeutic agents and a growing number of biological substances. Large clinical study-based chemotherapeutic protocols combined with biologicals have become an important component in (neo-) adjuvant therapy alongside surgery in solid cancers as well as radiation therapy in some instances. In recent years, monoclonal antibodies have entered the mainstream of cancer therapy. Their first use was as antagonists of oncogenic receptor tyrosine kinases, but today monoclonal antibodies have emerged as long-sought vehicles for the targeted delivery of potent chemotherapeutic agents and as powerful tools to manipulate anticancer immune responses. There is a growing number of FDA approved monoclonal antibodies and small molecules targeting specific types of cancer suggestive of the clinical relevance of this approach.Targeted cancer therapies , also referred to as personalized medicine, are being studied for use alone, in combination with other targeted therapies, and in combination with chemotherapy. The use of monoclonal antibodies in colorectal and gastric cancer for example have shown best outcome when combined with chemotherapy, even though single agent anti-EGFR antibodies seem to be active in particular setting of metastatic colorectal cancer patients. However, it is not well defined whether the addition of anti-VEGF - and anti-EGFR strategies to chemotherapy could improve outcome in those patients susceptible to colorectal cancer-related metastases resection. Among the most promising approaches to activating therapeutic antitumor immunity is the blockade of immune checkpoints, exemplified by the recently FDA-approved agent, Ipilimumab, an antibody that blocks the coinhibitory receptor CTLA-4. Capitalizing on the success of Ipilimumab, agents that target a second coinhibitory receptor, PD-1, or its ligand, PD-L1, are in clinical development. This section attempts to discuss recent progress of targeted agents and in tackling a more general target applicable to gastrointestinal cancer .
Collapse
|
13
|
Liang Y, Xu X, Wang T, Li Y, You W, Fu J, Liu Y, Jin S, Ji Q, Zhao W, Song Q, Li L, Hong T, Huang J, Lyu Z, Ye Q. The EGFR/miR-338-3p/EYA2 axis controls breast tumor growth and lung metastasis. Cell Death Dis 2017; 8:e2928. [PMID: 28703807 PMCID: PMC5550870 DOI: 10.1038/cddis.2017.325] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/30/2017] [Accepted: 06/12/2017] [Indexed: 02/07/2023]
Abstract
Dysregulation of the epidermal growth factor receptor (EGFR) promotes cancer cell growth, invasion and metastasis. However, its relevant downstream effectors are still limited. Here, we show that EGFR promotes breast tumor growth and metastasis by downregulating the tumor suppressor micoRNA-338-3p (miR-338-3p) and activating the EYA2 (EYA transcriptional coactivator and phosphatase 2) oncoprotein. EGFR represses miR-338-3p expression largely through HIF1α transcription factor. miR-338-3p inhibits EYA2 expression by binding to the 3'-untranslated region of EYA2. EGFR increases EYA2 expression via HIF1α repression of miR-338-3p. Through the miR-338-3p/EYA2 pathway, EGFR increases breast cancer cell growth, epithelial-to-mesenchymal transition, migration, invasion and lung metastasis in vitro and in a allograft tumor mouse model in vivo. In breast cancer patients, miR-338-3p expression negatively correlates with the expression of EGFR and EYA2, EGFR status positively associates with EYA2 expression, and miR-338-3p and EYA2 predict breast cancer lung metastasis when expressed in primary breast cancers. These data suggest that the miR-338-3p/EYA2 axis contributes to EGFR-mediated tumor growth and lung metastasis and that miR-338-3p activation or EYA2 inhibition or combination therapy targeting EGFR/miR-338-3p/EYA2 axis may be a promising way to treat patients with metastatic cancer.
Collapse
Affiliation(s)
- Yingchun Liang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Tao Wang
- Department of Oncology, 307 Hospital of People's Liberation Army, Beijing, China
| | - Ying Li
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Wenye You
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Jing Fu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Yang Liu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Thoracic Surgery, PLA General Hospital, Beijing, China
| | - Shuai Jin
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Thoracic Surgery, PLA General Hospital, Beijing, China
| | - Quanbo Ji
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
- Department of Orthopedics, PLA General Hospital, Beijing, China
| | - Wei Zhao
- Department of Oncology, The General Hospital of the PLA Rocket Force, Beijing, China
| | - Qi Song
- Department of Oncology, PLA General Hospital, Beijing, China
| | - Ling Li
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Tian Hong
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Junjian Huang
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| | - Zhaohui Lyu
- Department of Endocrinology, PLA General Hospital, Beijing, China
| | - Qinong Ye
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Collaborative Innovation Center for Cancer Medicine, Beijing, China
| |
Collapse
|
14
|
Weitsman G, Mitchell NJ, Evans R, Cheung A, Kalber TL, Bofinger R, Fruhwirth GO, Keppler M, Wright ZVF, Barber PR, Gordon P, de Koning T, Wulaningsih W, Sander K, Vojnovic B, Ameer-Beg S, Lythgoe M, Arnold JN, Årstad E, Festy F, Hailes HC, Tabor AB, Ng T. Detecting intratumoral heterogeneity of EGFR activity by liposome-based in vivo transfection of a fluorescent biosensor. Oncogene 2017; 36:3618-3628. [PMID: 28166195 PMCID: PMC5421598 DOI: 10.1038/onc.2016.522] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/12/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
Despite decades of research in the epidermal growth factor receptor (EGFR) signalling field, and many targeted anti-cancer drugs that have been tested clinically, the success rate for these agents in the clinic is low, particularly in terms of the improvement of overall survival. Intratumoral heterogeneity is proposed as a major mechanism underlying treatment failure of these molecule-targeted agents. Here we highlight the application of fluorescence lifetime microscopy (FLIM)-based biosensing to demonstrate intratumoral heterogeneity of EGFR activity. For sensing EGFR activity in cells, we used a genetically encoded CrkII-based biosensor which undergoes conformational changes upon tyrosine-221 phosphorylation by EGFR. We transfected this biosensor into EGFR-positive tumour cells using targeted lipopolyplexes bearing EGFR-binding peptides at their surfaces. In a murine model of basal-like breast cancer, we demonstrated a significant degree of intratumoral heterogeneity in EGFR activity, as well as the pharmacodynamic effect of a radionuclide-labeled EGFR inhibitor in situ. Furthermore, a significant correlation between high EGFR activity in tumour cells and macrophage-tumour cell proximity was found to in part account for the intratumoral heterogeneity in EGFR activity observed. The same effect of macrophage infiltrate on EGFR activation was also seen in a colorectal cancer xenograft. In contrast, a non-small cell lung cancer xenograft expressing a constitutively active EGFR conformational mutant exhibited macrophage proximity-independent EGFR activity. Our study validates the use of this methodology to monitor therapeutic response in terms of EGFR activity. In addition, we found iNOS gene induction in macrophages that are cultured in tumour cell-conditioned media as well as an iNOS activity-dependent increase in EGFR activity in tumour cells. These findings point towards an immune microenvironment-mediated regulation that gives rise to the observed intratumoral heterogeneity of EGFR signalling activity in tumour cells in vivo.
Collapse
Affiliation(s)
- G Weitsman
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - N J Mitchell
- Department of Chemistry, University College London, London, UK
| | - R Evans
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - A Cheung
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
- Breast Cancer Now Research Unit, King’s College London, London, UK
| | - T L Kalber
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - R Bofinger
- Department of Chemistry, University College London, London, UK
| | - G O Fruhwirth
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - M Keppler
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - Z V F Wright
- Department of Chemistry, University College London, London, UK
| | - P R Barber
- Gray Laboratories, Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - P Gordon
- Breast Cancer Now Research Unit, King’s College London, London, UK
| | - T de Koning
- Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - W Wulaningsih
- Cancer Epidemiology Group, Division of Cancer Studies, King’s College London, London, UK
| | - K Sander
- Institute of Nuclear Medicine, University College London, London, UK
| | - B Vojnovic
- Gray Laboratories, Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - S Ameer-Beg
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - M Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - J N Arnold
- Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - E Årstad
- Institute of Nuclear Medicine, University College London, London, UK
| | - F Festy
- King’s College London Dental Institute, Tissue Engineering and Biophotonics, Guy’s Hospital Campus, London, UK
| | - H C Hailes
- Department of Chemistry, University College London, London, UK
| | - A B Tabor
- Department of Chemistry, University College London, London, UK
| | - T Ng
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
- Breast Cancer Now Research Unit, King’s College London, London, UK
- UCL Cancer Institute, Paul O’Gorman Building, University College London, London, UK
| |
Collapse
|
15
|
Dual PI3K and Wnt pathway inhibition is a synergistic combination against triple negative breast cancer. NPJ Breast Cancer 2017. [PMID: 28649657 PMCID: PMC5460220 DOI: 10.1038/s41523-017-0016-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Triple negative breast cancer accounts for 15–20% of all breast cancer cases, but despite its lower incidence, contributes to a disproportionately higher rate of mortality. As there are currently no Food and Drug Administration-approved targeted agents for triple negative breast cancer, we embarked on a genomic-guided effort to identify novel targeted modalities. Analyses by our group and The Cancer Genome Atlas have identified activation of the PI3K-pathway in the majority of triple negative breast cancers. As single agent therapy is commonly subject to resistance, we investigated the use of combination therapy against compensatory pathways. Herein, we demonstrate that pan-PI3K inhibition in triple negative breast cancers results in marked activation of the Wnt-pathway. Using the combination of two inhibitors currently in clinical trial as single agents, buparlisib(pan-PI3K) and WNT974(WNT-pathway), we demonstrate significant in vitro and in vivo synergy against triple negative breast cancer cell lines and xenografts. Taken together, these observations provide a strong rationale for testing dual targeting of the PI3K and WNT-pathways in clinical trials. Two experimental drugs currently in clinical trials as single agents might work better together against triple negative breast cancer. Jeffrey Solzak and Milan Radovich, and colleagues from the Indiana University School of Medicine in Indianapolis, USA, analyzed gene expression data from cancerous and healthy breast tissues and from published genomic datasets. They found the PI3K pathway consistently activated in the majority of breast cancers that test negative for the three most common receptors known to fuel breast tumor growth. Inhibiting PI3K in these cells led to a marked increase in the activity of another critical pathway called Wnt, an observation that prompted the researchers to test a combination of a PI3K inhibitor, buparlisib, and a Wnt inhibitor, WNT974, in cell lines and xenograft models. Tumor cells shrunk more and mice lived longer than treatment with either agent alone.
Collapse
|
16
|
Matsuda N, Lim B, Wang X, Ueno NT. Early clinical development of epidermal growth factor receptor targeted therapy in breast cancer. Expert Opin Investig Drugs 2017; 26:463-479. [PMID: 28271910 PMCID: PMC5826640 DOI: 10.1080/13543784.2017.1299707] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 02/22/2017] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Epidermal growth factor receptor (EGFR) targeted treatment has been evaluated but has not shown a clear clinical benefit for breast cancer. This review article aims to consider the knowledge of the biological background of EGFR pathways in dissecting clinical studies of EGFR targeted treatment in breast cancer. Areas covered: This review focuses on the role of the EGFR pathway and the investigational drugs that target EGFR for breast cancer. Expert opinion: Recent studies have indicated that EGFR targeted therapy for breast cancer has some promising effects for patients with triple-negative breast cancer, basal-like breast cancer, and inflammatory breast cancer. However, predictive and prognostic biomarkers for EGFR targeted therapy have not been identified. The overexpression or amplification of EGFR itself may not be the true factor of induction of the canonical pathway as an oncogenic driver of breast cancer. Instead, downstream, non-canonical pathways related to EGFR may contribute to some aspects of the biological behavior of breast cancer; therefore, the blockade of the receptor could result in sufficient suppression of downstream pathways to inhibit the aggressive behavior of breast cancer. Mechanistic studies to investigate the dynamic interaction between the EGFR pathway and non-canonical pathways are warranted.
Collapse
Affiliation(s)
- Naoko Matsuda
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bora Lim
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaoping Wang
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naoto T. Ueno
- Section of Translational Breast Cancer Research, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Morgan Welch Inflammatory Breast Cancer Research Program and Clinic, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
17
|
EGFR-independent Elk1/CIP2A signalling mediates apoptotic effect of an erlotinib derivative TD52 in triple-negative breast cancer cells. Eur J Cancer 2017; 72:112-123. [DOI: 10.1016/j.ejca.2016.11.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 10/27/2016] [Accepted: 11/15/2016] [Indexed: 12/23/2022]
|
18
|
Bernad IP, Trufero JM, Urquizu LC, Pazo Cid RA, de Miguel AC, Agustin MJ, Lanzuela M, Antón A. Activity of weekly paclitaxel-cetuximab chemotherapy in unselected patients with recurrent/metastatic head and neck squamous cell carcinoma: prognostic factors. Clin Transl Oncol 2017; 19:769-776. [PMID: 28120324 DOI: 10.1007/s12094-016-1604-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 12/19/2016] [Indexed: 01/30/2023]
Abstract
BACKGROUND Standard treatment for recurrent/metastatic head and neck squamous cell carcinoma (RM-SCCHN) is based in on platinum and cetuximab combination therapy. Unfortunately, not all patients are candidates to receive platinum-based treatment, because of different conditions as comorbidity and poor performance status. Weekly paclitaxel and cetuximab (WPC) is an active therapeutic alternative, based on a phase II study, with less toxicity. Our main objective is to confirm its activity in unselected patients, mostly unfit for aggressive therapies, analysing also some clinically relevant prognostic factors (PFs). METHODS Retrospective data was collected for RM-SCCHN patients, treated at our institution between January 2008 and July 2014 with weekly paclitaxel (80 mg/m2) and cetuximab (400/250 mg/m2). RESULTS 148 patients were treated. The objective response rate (OR) was as follows: 13 patients (8.78%) complete response (CR); 57 patients (38.51%) partial response (PR) and 30 patients (20.3%) stable disease (SD). Median overall survival (OS) was 10 months (95% CI 8.31-11.69) and median progression free survival (PFS) was 7 months (95% CI 5.88-8.12). Response to treatment showed independent prognosis relevance as PF in multivariate analysis for PFS and OS. Furthermore, decline in serum magnesium during the treatment was also an independent PF for OS. CONCLUSIONS WPC activity was confirmed as a useful therapy on real-life unselected RM-SCCHN patients, with similar benefit to that obtained in the phase II study, and comparable to platinum and cetuximab based treatment, confirming its value in unfit patients. In addition to treatment response, a change in serum magnesium values during treatment was proved as independent PF on OS.
Collapse
Affiliation(s)
- I Pajares Bernad
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain.
| | - J Martínez Trufero
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain
| | - L Calera Urquizu
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain
| | - R A Pazo Cid
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain
| | - A Cebollero de Miguel
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain
| | - M J Agustin
- Pharmacy Department, Miguel Servet University Hospital, Saragossa, Spain
| | - M Lanzuela
- Radiotherapy Department, Miguel Servet University Hospital, Saragossa, Spain
| | - A Antón
- Medical Oncology Department, Miguel Servet University Hospital, 1-3 Isabel la Catolica Avenue, 50009, Saragossa, Spain
| |
Collapse
|
19
|
Wang YL, Overstreet AM, Chen MS, Wang J, Zhao HJ, Ho PC, Smith M, Wang SC. Combined inhibition of EGFR and c-ABL suppresses the growth of triple-negative breast cancer growth through inhibition of HOTAIR. Oncotarget 2016; 6:11150-61. [PMID: 25883211 PMCID: PMC4484446 DOI: 10.18632/oncotarget.3441] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 02/26/2015] [Indexed: 01/19/2023] Open
Abstract
Triple-negative breast cancer (TNBC) does not express conventional therapeutic targets and is the only type of malignant breast cancer for which no designated FDA-approved targeted therapy is available. Although overexpression of epidermal growth factor receptor (EGFR) is frequently found in TNBC, the therapeutic effect of EGFR inhibitors in TNBC has been underwhelming. Here we show that co-treatment with clinically validated inhibitors of c-ABL (imatinib) and EGFR (lapatinib) results in synergistic growth inhibition in TNBC cells. The dual treatment leads to synergistic repression of the long non-coding RNA (lncRNA) HOTAIR (HOX Antisense Intergenic RNA). HOTAIR has been known to induce tumor growth and metastasis in breast cancer. Depleting HOTAIR alone phenocopies the dual treatment in growth suppression. We show that expression of HOTAIR is regulated by β-catenin through a LEF1/TCF4-binding site. The dual treatment blocks nuclear expression of β-catenin and prevents its recruitment to the HOTAIR promoter. Consistently, forced expression of β-catenin rescued HOTAIR expression and cell viability in the presence of both drugs. Upregulation of HOTAIR is associated with TNBC in cell lines and a cohort of primary tumors. This study elucidates a previously unidentified mechanism in TNBC linking signaling with lncRNA regulation which may be exploited for therapeutic gain.
Collapse
Affiliation(s)
- Yuan-Liang Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Anne-Marie Overstreet
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Min-Shan Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jiang Wang
- Department of Pathology & Lab Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hua-Jun Zhao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,School of Pharmacy, Zhejiang Chinese Medical University, Zhejiang, China
| | - Po-Chun Ho
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Molly Smith
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Cancer and Cell Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Shao-Chun Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Cancer and Cell Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
20
|
Kim J, Won Y, Goh SH, Choi Y. A redox-responsive theranostic agent for target-specific fluorescence imaging and photodynamic therapy of EGFR-overexpressing triple-negative breast cancers. J Mater Chem B 2016; 4:6787-6790. [DOI: 10.1039/c6tb01519k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A redox-responsive specific theranostic agent for target-cell-specific activatable fluorescence imaging and photodynamic therapy of triple-negative breast cancers.
Collapse
Affiliation(s)
- Jisu Kim
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
| | - Youngjae Won
- Medical Device Development Center
- Osong Medical Innovation Foundation
- Republic of Korea
| | - Sung-Ho Goh
- Precision Medicine Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
| | - Yongdoo Choi
- Molecular Imaging & Therapy Branch
- National Cancer Center
- Goyang-si
- Republic of Korea
- Precision Medicine Branch
| |
Collapse
|
21
|
Nagaya T, Sato K, Harada T, Nakamura Y, Choyke PL, Kobayashi H. Near Infrared Photoimmunotherapy Targeting EGFR Positive Triple Negative Breast Cancer: Optimizing the Conjugate-Light Regimen. PLoS One 2015; 10:e0136829. [PMID: 26313651 PMCID: PMC4552472 DOI: 10.1371/journal.pone.0136829] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/10/2015] [Indexed: 01/22/2023] Open
Abstract
Aim Triple-negative breast cancer (TNBC) is considered one of the most aggressive subtypes of breast cancer. Near infrared photoimmunotherapy (NIR-PIT) is a cancer treatment that employs an antibody-photosensitizer conjugate (APC) followed by exposure of NIR light for activating selective cytotoxicity on targeted cancer cells and may have application to TNBC. In order to minimize the dose of APC while maximizing the therapeutic effects, dosing of the APC and NIR light need to be optimized. In this study, we investigate in vitro and in vivo efficacy of cetuximab (cet)-IR700 NIR-PIT on two breast cancer models MDAMB231 (TNBC, EGFR moderate) and MDAMB468 (TNBC, EGFR high) cell lines, and demonstrate a method to optimize the dosing APC and NIR light. Method After validating in vitro cell-specific cytotoxicity, NIR-PIT therapeutic effects were investigated in mouse models using cell lines derived from TNBC tumors. Tumor-bearing mice were separated into 4 groups for the following treatments: (1) no treatment (control); (2) 300 μg of cet-IR700 i.v., (APC i.v. only); (3) NIR light exposure only, NIR light was administered at 50 J/cm2 on day 1 and 100 J/cm2 on day 2 (NIR light only); (4) 300 μg of cet-IR700 i.v., NIR light was administered at 50 J/cm2 on day 1 after injection and 100 J/cm2 of light on day 2 after injection (one shot NIR-PIT). To compare different treatment regimens with a fixed dose of APC, we added the following treatments (5) 100 μg of cet-IR700 i.v., NIR light administered at 50 J/cm2 on day 1 and 50 μg of cet-IR700 i.v. immediately after NIR-PIT, then NIR light was administered at 100 J/cm2 on day 2, which were performed two times every week (“two split” NIR-PIT) and (6) 100 μg of cet-IR700 i.v., NIR light was administered at 50 J/cm2 on day 1 and 100 J/cm2 on day 2, which were performed three times per week (“three split” NIR-PIT). Result Both specific binding and NIR-PIT effects were greater with MDAMB468 than MDAMB231 cells in vitro. Tumor accumulation of cet-IR700 in MDAMB468 tumors was significantly higher (p < 0.05) than in MDAMB231 tumors in vivo. Tumor growth and survival of MDAMB231 tumor bearing mice was significantly lower in the NIR-PIT treatment group (p < 0.05). In MDAMB468 bearing mice, tumor growth and survival was significantly improved in the NIR-PIT treatment groups in all treatment regimens (one shot NIR-PIT; p < 0.05, “two split” NIR-PIT; p < 0.01, “three split” NIR-PIT; p < 0.001) compared with control groups. Conclusion NIR-PIT for TNBC was effective regardless of expression of EGFR, however, greater cell killing was shown with higher EGFR expression tumor in vitro. In all treatment regimens, NIR-PIT suppressed tumor growth, resulting in significantly prolonged survival that further improved by splitting the APC dose and using repeated light exposures.
Collapse
Affiliation(s)
- Tadanobu Nagaya
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Kazuhide Sato
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Toshiko Harada
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Yuko Nakamura
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Peter L Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
22
|
Crozier JA, Advani PP, LaPlant B, Hobday T, Jaslowski AJ, Moreno-Aspitia A, Perez EA. N0436 (Alliance): A Phase II Trial of Irinotecan With Cetuximab in Patients With Metastatic Breast Cancer Previously Exposed to Anthracycline and/or Taxane-Containing Therapy. Clin Breast Cancer 2015; 16:23-30. [PMID: 26381420 DOI: 10.1016/j.clbc.2015.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/10/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Irinotecan has a 20% to 25% response rate (RR) in patients with previously treated metastatic breast cancer (MBC). Epidermal growth factor receptor (EGFR) is overexpressed in some MBC, especially in triple-negative breast cancer (TNBC). Cetuximab is a monoclonal antibody against EGFR with additive preclinical activity with irinotecan. PATIENTS AND METHODS We report a 1-stage phase II study on MBC, measurable disease, and previous anthracycline and/or taxane therapy. Patients received cetuximab 400 mg/m(2) on day 1 cycle 1 then 250 mg/m(2) weekly thereafter and irinotecan 80 mg/m(2) on days 1 and 8 of each 21-day cycle. The primary end point was overall RR (ORR) according to Response Evaluation Criteria in Solid Tumors criteria (version 1.1). RESULTS Of 19 eligible patients enrolled from February to September 2006, 14 patients (74%) had visceral disease, seven patients (37%) were hormone receptor-positive, two patients (11%) HER2-positive, and 11 patients (58%) were triple-negative. Patients received a median of 2 cycles (range, 1-37). Confirmed ORR was 11% (95% confidence interval [CI], 1%-33%), with 1 partial response and 1 complete response. One patient had stable disease for 8 months. RR for TNBC versus non-TNBC was 18% versus 0% (P = .49). Median time to progression was 1.4 months (95% CI, 1.0-2.2) and median overall survival was 9.4 months (95% CI, 2.8-16.1). Twelve patients had disease progression within 2 cycles during therapy. Because of a low RR and rapid disease progression, the study leadership decided to close the trial early. CONCLUSION The tolerability of the combination of cetuximab and irinotecan is acceptable but demonstrated low overall activity. Potentially promising results were noted in patients with TNBC and further studies of these patients might be considered.
Collapse
Affiliation(s)
| | - Pooja P Advani
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL
| | - Betsy LaPlant
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN
| | - Timothy Hobday
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | | | | | - Edith A Perez
- Division of Hematology/Oncology, Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
23
|
Wang Y, Zhang C, Zhang Y, Fang H, Min C, Zhu S, Yuan XC. Investigation of phase SPR biosensor for efficient targeted drug screening with high sensitivity and stability. SENSORS AND ACTUATORS B: CHEMICAL 2015; 209:313-322. [DOI: 10.1016/j.snb.2014.11.134] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/09/2024]
|
24
|
Tural D, Akar E, Mutlu H, Kilickap S. P95 HER2 fragments and breast cancer outcome. Expert Rev Anticancer Ther 2014; 14:1089-96. [DOI: 10.1586/14737140.2014.929946] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Wang SC. PCNA: a silent housekeeper or a potential therapeutic target? Trends Pharmacol Sci 2014; 35:178-86. [PMID: 24655521 DOI: 10.1016/j.tips.2014.02.004] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 02/17/2014] [Accepted: 02/18/2014] [Indexed: 11/18/2022]
Abstract
Proliferating cell nuclear antigen (PCNA) is known as a molecular marker for proliferation given its role in replication. Three identical molecules of PCNA form a molecular sliding clamp around the DNA double helix. This provides an essential platform on which multiple proteins are dynamically recruited and coordinately regulated. Over the past decade, new research has provided a deeper comprehension of PCNA as a coordinator of essential cellular functions for cell growth, death, and maintenance. Although the biology of PCNA in proliferation has been comprehensively reviewed, research progress in unveiling the potential of targeting PCNA for disease treatment has not been systematically discussed. Here we briefly summarize the basic structural and functional characteristics of PCNA, and then discuss new developments in its protein interactions, trimer formation, and signaling regulation that open the door to possible therapeutic targeting of PCNA.
Collapse
Affiliation(s)
- Shao-Chun Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
26
|
Abstract
Within the past 2 years, four separate groups have reported marked improvement in relapse-free survival when trastuzumab was added to adjuvant chemotherapy in patients with HER2-overexpressing breast cancer. These results add further credence to the relevance of this receptor as a tumor target. Despite the significant benefits observed in early and advanced HER2-positive breast cancer, overexpression of the receptor is still associated with a poorer prognosis and an increased risk of disease relapse, even in patients with primary operable disease. Besides cytotoxic chemotherapy, and possibly hormonal therapy, patients whose tumors exhibit resistance to trastuzumab have few molecular-targeted options available. Recently, lapatinib, a small molecule dual inhibitor of both HER2 and EGF receptors, has been developed to expand the options for treating HER-positive breast cancer.
Collapse
Affiliation(s)
- Gerald M Higa
- Mary Babb Randolph Cancer Center, School of Pharmacy, West Virginia University, Morgantown, WV 26506, USA.
| | | |
Collapse
|
27
|
Tong L, Yang XX, Liu MF, Yao GY, Dong JY, Ye CS, Li M. Mutational analysis of key EGFR pathway genes in Chinese breast cancer patients. Asian Pac J Cancer Prev 2013; 13:5599-603. [PMID: 23317280 DOI: 10.7314/apjcp.2012.13.11.5599] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) is a potential therapeutic target for breast cancer; however, its use does not lead to a marked clinical response. Studies of non-small cell lung cancer and colorectal cancer showed that mutations of genes in the PIK3CA/AKT and RAS/RAF/MEK pathways, two major signalling cascades downstream of EGFR, might predict resistance to EGFR-targeted agents. Therefore, we examined the frequencies of mutations in these key EGFR pathway genes in Chinese breast cancer patients. METHODS We used a high-throughput mass-spectrometric based cancer gene mutation profiling platform to detect 22 mutations of the PIK3CA, AKT1, BRAF, EGFR, HRAS, and KRAS genes in 120 Chinese women with breast cancer. RESULTS Thirteen mutations were detected in 12 (10%) of the samples, all of which were invasive ductal carcinomas (two stage I, six stage II, three stage III, and one stage IV). These included one mutation (0.83%) in the EGFR gene (rs121913445-rs121913432), three (2.5%) in the KRAS gene (rs121913530, rs112445441), and nine (7.5%) in the PIK3CA gene (rs121913273, rs104886003, and rs121913279). No mutations were found in the AKT1, BRAF, and HRAS genes. Six (27.3%) of the 22 genotyping assays caused mutations in at least one sample and three (50%) of the six assays queried were found to be mutated more than once. CONCLUSIONS Mutations in the EGFR pathway occurred in a small fraction of Chinese breast cancers. However, therapeutics targeting these potential predictive markers should be investigated in depth, especially in Oriental populations.
Collapse
Affiliation(s)
- Lin Tong
- School of Biotechnology, Breast Center Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Shapira I, Lee A, Vora R, Budman DR. P53 mutations in triple negative breast cancer upregulate endosomal recycling of epidermal growth factor receptor (EGFR) increasing its oncogenic potency. Crit Rev Oncol Hematol 2013; 88:284-92. [PMID: 23755891 DOI: 10.1016/j.critrevonc.2013.05.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 04/03/2013] [Accepted: 05/02/2013] [Indexed: 01/03/2023] Open
Abstract
There is no available targeted therapy for triple-negative or its more aggressive subtype, basal-like breast cancer. Multiple therapeutic strategies based on translational knowledge have not improved the treatment options for triple negative patients. As understanding of molecular pathways that drive tumor development is rapidly increasing, it is imperative to adapt our treatment strategies to perturbations in molecular pathways driving the malignant process. Basal-like breast cancers over-express EGFR (without mutations or EGFR gene amplifications) and have p53 mutations. While EGFR drives the malignant behavior in triple negative breast cancer (TNBC), anti-EGFR therapies have fallen short of the expected results in clinical trials. Here we bring evidence that the less than optimal results of the anti-EGFR therapies may be explained in part by the increased potency of the EGFR signaling due to increased endosomal recycling. The functional connection between EGFR and endosomal trafficking in TNBC is mutant p53 found in the most aggressive forms of TNBC. Mutant p53 acquires oncogenic functions and binds p63 protein, a member of p53 family with tumor suppressor activities. In the absence of functional p63 there is an upregulation of endosomal recycling EGFR and integrin to the membrane with increased proinvasive abilities of cancer cells. Blocking endosomal trafficking combined with anti-EGFR treatments may result in better clinical outcomes in TNBC.
Collapse
Affiliation(s)
- Iuliana Shapira
- Monter Cancer Center, Don Monti Division of Oncology, Division of Hematology, Hofstra North Shore Long Island Jewish School of Medicine, United States.
| | | | | | | |
Collapse
|
29
|
Abstract
INTRODUCTION Dasatinib is a novel tyrosine kinase inhibitor with activity against the Src family kinases. The Src pathway is active in breast cancer and has been shown to promote cell proliferation, invasiveness, and metastases. Preclinical data support the use of dasatinib to inhibit breast cancer cell growth, modulate hormone and EGFR signaling, and alter cell migration. AREAS COVERED This article details the preclinical data supporting dasatinib's use in breast cancer treatment. Additionally, Phase I/II studies that have tested dasatinib as a single agent and in combination with chemotherapy and antihormonal agents are discussed. EXPERT OPINION Phase I/II studies to date have shown only limited responses to dasatinib among breast cancer patients. Dasatinib has no current role in the treatment of breast cancer, and its future is uncertain. No specific subset of patients has been identified which preferentially responds to treatment. Studies are under way to identify specific molecular markers that might predict response to dasatinib.
Collapse
Affiliation(s)
- Kevin S Scher
- City of Hope Comprehensive Cancer Center, 1500 Duarte Road, Duarte, CA 91010-3012, USA
| | | |
Collapse
|
30
|
Role of epidermal growth factor receptor in breast cancer. Breast Cancer Res Treat 2012; 136:331-45. [PMID: 23073759 DOI: 10.1007/s10549-012-2289-9] [Citation(s) in RCA: 506] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 10/03/2012] [Indexed: 02/07/2023]
Abstract
Decades of research in molecular oncology have brought about promising new therapies which are designed to target specific molecules which promote tumor growth and survival. The epidermal growth factor receptor (EGFR) is one of the first identified important targets of these novel antitumor agents. Approximately half of cases of triple-negative breast cancer (TNBC) and inflammatory breast cancer (IBC) overexpress EGFR. Thus, EGFR inhibitors for treatment of breast cancer have been evaluated in several studies. However, results so far have been disappointing. One of the reasons for these unexpected results is the lack of biomarkers for predicting which patients are most likely to respond to EGFR inhibitors. Recent studies have shown that EGFR and its downstream pathway regulate epithelial-mesenchymal transition, migration, and tumor invasion and that high EGFR expression is an independent predictor of poor prognosis in IBC. Further, recent studies have shown that targeting EGFR enhances the chemosensitivity of TNBC cells by rewiring apoptotic signaling networks in TNBC. These studies indicate that EGFR-targeted therapy might have a promising role in TNBC and IBC. Further studies of the role of EGFR in TNBC and IBC are needed to better understand the best way to use EGFR-targeted therapy-e.g., as a chemosensitizer or to prevent metastases-to treat these aggressive diseases.
Collapse
|
31
|
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
|
32
|
Flotillins as regulators of ErbB2 levels in breast cancer. Oncogene 2012; 32:3443-51. [PMID: 22869152 DOI: 10.1038/onc.2012.357] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 06/29/2012] [Accepted: 06/30/2012] [Indexed: 01/07/2023]
Abstract
Amplification and overexpression of the receptor tyrosine kinase ErbB2 occur in up to 30% of human breast cancers, and high ErbB2 levels are correlated with poor prognosis for breast cancer patients. In contrast to the epithelial growth factor receptor (ErbB1), ErbB2 is not downregulated by ligand-induced mechanisms. Here we show that flotillins are involved in the stabilization of ErbB2 at the plasma membrane. In SKBR3 breast cancer cells and breast cancer tissue, a positive correlation between flotillin and ErbB2 expression levels could be demonstrated. Moreover, the tissue microarray analyses of biopsies from 194 patients diagnosed with carcinomas of the breast showed that flotillin-2 emerged as a potential predictor of prognosis in breast cancer. Depletion of flotillin-1 and flotillin-2 leads to internalization and degradation of ErbB2. Furthermore, flotillin-1 and -2 were found to be in a molecular complex with ErbB2 and Hsp90. The depletion of one of these proteins results in disruption of this complex, followed by destabilization of ErbB2 at the membrane, and its internalization and degradation. As a consequence, ErbB2-triggered downstream signalling is inhibited. Our data demonstrate a novel mechanism for interfering with ErbB2 signalling, which potentially can have clinical impact.
Collapse
|
33
|
Metronomic chemotherapy combined with bevacizumab and erlotinib in patients with metastatic HER2-negative breast cancer: clinical and biological activity. Clin Breast Cancer 2012; 12:207-14. [PMID: 22520733 DOI: 10.1016/j.clbc.2012.03.008] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 03/09/2012] [Accepted: 03/16/2012] [Indexed: 01/15/2023]
Abstract
UNLABELLED The aim of this study was to determine the safety and efficacy of metronomic chemotherapy combined with targeted drugs in patients with metastatic breast cancer (MBC). We included 26 untreated patients with HER2-negative (HER-) MBC and poor hormone receptor expression. The analysis of the results suggests that the metronomic chemotherapy combined with bevacizumab and erlotinib is effective and well tolerated. BACKGROUND The object of this study was to evaluate the safety and efficacy of metronomic chemotherapy in combination with bevacizumab and erlotinib in patients with HER2-negative (HER2(-)) metastatic breast cancer (MBC) and poor hormone receptor expression. PATIENTS AND METHODS Patients with untreated MBC were candidates to receive metronomic oral capecitabine (500 mg thrice daily) and cyclophosphamide (50 mg daily) plus bevacizumab (15 mg/kg every 3 weeks) and erlotinib (100 mg daily). RESULTS Of 24 patients assessable for response, we observed 1 complete response (CR, 4%), 14 partial responses (58%), 5 patients with stable disease greater than 9 weeks' duration (SD, 21%), and 1 patient (4%) with early progression of disease. The overall clinical benefit (CB) (CR + partial response + SD > 24 weeks) was 75% (95% confidence interval [CI], 53%-90%). Median time to progression was 43 weeks (95% CI, 21-69). Patients with low levels of circulating endothelial progenitors (CEPs) at baseline had a significantly improved progression-free survival (PFS). Toxicity was generally mild. Grade 3 toxicity included diarrhea (n = 1), thrombosis (n = 1), and hypertension (n = 2). Grade 2 adverse events included diarrhea (n = 5), hand-foot syndrome (n = 13), and hypertension (n = 4). CONCLUSION Treatment with metronomic chemotherapy in combination with bevacizumab and erlotinib was effective in HER2(-), estrogen receptor (ER)- and progesterone receptor (PR)-poor advanced breast cancer.
Collapse
|
34
|
Pichinuk E, Benhar I, Jacobi O, Chalik M, Weiss L, Ziv R, Sympson C, Karwa A, Smorodinsky NI, Rubinstein DB, Wreschner DH. Antibody targeting of cell-bound MUC1 SEA domain kills tumor cells. Cancer Res 2012; 72:3324-36. [PMID: 22507854 DOI: 10.1158/0008-5472.can-12-0067] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The cell-surface glycoprotein MUC1 is a particularly appealing target for antibody targeting, being selectively overexpressed in many types of cancers and a high proportion of cancer stem-like cells. However the occurrence of MUC1 cleavage, which leads to the release of the extracellular α subunit into the circulation where it can sequester many anti-MUC1 antibodies, renders the target problematic to some degree. To address this issue, we generated a set of unique MUC1 monoclonal antibodies that target a region termed the SEA domain that remains tethered to the cell surface after MUC1 cleavage. In breast cancer cell populations, these antibodies bound the cancer cells with high picomolar affinity. Starting with a partially humanized antibody, DMB5F3, we created a recombinant chimeric antibody that bound a panel of MUC1+ cancer cells with higher affinities relative to cetuximab (anti-EGFR1) or tratuzumab (anti-erbB2) control antibodies. DMB5F3 internalization from the cell surface occurred in an efficient temperature-dependent manner. Linkage to toxin rendered these DMB5F3 antibodies to be cytotoxic against MUC1+ cancer cells at low picomolar concentrations. Our findings show that high-affinity antibodies to cell-bound MUC1 SEA domain exert specific cytotoxicity against cancer cells, and they point to the SEA domain as a potential immunogen to generate MUC1 vaccines.
Collapse
Affiliation(s)
- Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Inoue S, Patil R, Portilla-Arias J, Ding H, Konda B, Espinoza A, Mongayt D, Markman JL, Elramsisy A, Phillips HW, Black KL, Holler E, Ljubimova JY. Nanobiopolymer for direct targeting and inhibition of EGFR expression in triple negative breast cancer. PLoS One 2012; 7:e31070. [PMID: 22355336 PMCID: PMC3280290 DOI: 10.1371/journal.pone.0031070] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Accepted: 01/02/2012] [Indexed: 12/31/2022] Open
Abstract
Treatment options for triple negative breast cancer (TNBC) are generally limited to cytotoxic chemotherapy. Recently, anti-epidermal growth factor receptor (EGFR) therapy has been introduced for TNBC patients. We engineered a novel nanobioconjugate based on a poly(β-L-malic acid) (PMLA) nanoplatform for TNBC treatment. The nanobioconjugate carries anti-tumor nucleosome-specific monoclonal antibody (mAb) 2C5 to target breast cancer cells, anti-mouse transferrin receptor (TfR) antibody for drug delivery through the host endothelial system, and Morpholino antisense oligonucleotide (AON) to inhibit EGFR synthesis. The nanobioconjugates variants were: (1) P (BioPolymer) with AON, 2C5 and anti-TfR for tumor endothelial and cancer cell targeting, and EGFR suppression (P/AON/2C5/TfR), and (2) P with AON and 2C5 (P/AON/2C5). Controls included (3) P with 2C5 but without AON (P/2C5), (4) PBS, and (5) P with PEG and leucine ester (LOEt) for endosomal escape (P/mPEG/LOEt). Drugs were injected intravenously to MDA-MB-468 TNBC bearing mice. Tissue accumulation of injected nanobioconjugates labeled with Alexa Fluor 680 was examined by Xenogen IVIS 200 (live imaging) and confocal microscopy of tissue sections. Levels of EGFR, phosphorylated and total Akt in tumor samples were detected by western blotting. In vitro western blot showed that the leading nanobioconjugate P/AON/2C5/TfR inhibited EGFR synthesis significantly better than naked AON. In vivo imaging revealed that 2C5 increased drug-tumor accumulation. Significant tumor growth inhibition was observed in mice treated with the lead nanobioconjugate (1) [P = 0.03 vs. controls; P<0.05 vs. nanobioconjugate variant (2)]. Lead nanobioconjugate (1) also showed stronger inhibition of EGFR expression and Akt phosphorylation than other treatments. Treatment of TNBC with the new nanobioconjugate results in tumor growth arrest by inhibiting EGFR and its downstream signaling intermediate, phosphorylated Akt. The nanobioconjugate represents a new generation of nanodrugs for treatment of TNBC.
Collapse
Affiliation(s)
- Satoshi Inoue
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Rameshwar Patil
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jose Portilla-Arias
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Hui Ding
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bindu Konda
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andres Espinoza
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Dmitriy Mongayt
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, Massachusetts, United States of America
| | - Janet L. Markman
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Adam Elramsisy
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - H. Westley Phillips
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Keith L. Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Eggehard Holler
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Julia Y. Ljubimova
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
Jacot W, Lopez-Crapez E, Thezenas S, Senal R, Fina F, Bibeau F, Romieu G, Lamy PJ. Lack of EGFR-activating mutations in European patients with triple-negative breast cancer could emphasise geographic and ethnic variations in breast cancer mutation profiles. Breast Cancer Res 2011; 13:R133. [PMID: 22192147 PMCID: PMC3326575 DOI: 10.1186/bcr3079] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Revised: 12/13/2011] [Accepted: 12/22/2011] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Triple-negative breast cancers (TNBCs) are characterised by lack of expression of hormone receptors and epidermal growth factor receptor 2 (HER-2). As they frequently express epidermal growth factor receptors (EGFRs), anti-EGFR therapies are currently assessed for this breast cancer subtype as an alternative to treatments that target HER-2 or hormone receptors. Recently, EGFR-activating mutations have been reported in TNBC specimens in an East Asian population. Because variations in the frequency of EGFR-activating mutations in East Asians and other patients with lung cancer have been described, we evaluated the EGFR mutational profile in tumour samples from European patients with TNBC. METHODS We selected from a DNA tumour bank 229 DNA samples isolated from frozen, histologically proven and macrodissected invasive TNBC specimens from European patients. PCR and high-resolution melting (HRM) analyses were used to detect mutations in exons 19 and 21 of EGFR. The results were then confirmed by bidirectional sequencing of all samples. RESULTS HRM analysis allowed the detection of three EGFR exon 21 mutations, but no exon 19 mutations. There was 100% concordance between the HRM and sequencing results. The three patients with EGFR exon 21 abnormal HRM profiles harboured the rare R836R SNP, but no EGFR-activating mutation was identified. CONCLUSIONS This study highlights variations in the prevalence of EGFR mutations in TNBC. These variations have crucial implications for the design of clinical trials involving anti-EGFR treatments in TNBC and for identifying the potential target population.
Collapse
Affiliation(s)
- William Jacot
- Département d'Oncologie Médicale, CRLC Val d'Aurelle-Paul Lamarque, 208, rue des Apothicaires, F-34298, France.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Triple-negative breast cancer, characterized by tumors that do not express estrogen receptor (ER), progesterone receptor (PR), or HER-2 genes, represents an important clinical challenge because these cancers do not respond to endocrine therapy or other available targeted agents. The metastatic potential in triple-negative breast cancer is similar to that of other breast cancer subtypes, but these tumors are associated with a shorter median time to relapse and death. One important goal is therefore the identification of prognostic factors and markers to reliably select high and low risk subsets of patients with triple-negative disease for different treatment approaches of subtypes with differential responsiveness to specific agents. However, a reliable prognostic marker has been elusive, and markers have been inconsistently useful. For example, epidermal growth factor receptor (EGFR) has been studied, but there is still a lack of agreement on a standard assay or cutoff for EGFR expression levels with respect to prognosis. Similarly, because triple-negative status is sometimes used as a surrogate for basal-like breast cancer, specific basal markers have been explored. Indeed, trials designed to accrue patients with basal-like breast cancer using ER/PR and HER-2 negativity may provide only an approximation of the triple-negative population and are sometimes reanalyzed using more specific indicators like CK 5/6, EGFR status, and others, again marred by discordances. Chemotherapy remains the mainstay of treatment of triple-negative breast cancer, but important limitations still need to be overcome in the next few years if any significant clinical strides are to be made. Current treatment strategies for triple-negative disease include anthracyclines, taxanes, ixabepilone, platinum agents, and biologic agents. More recently, EGFR inhibition has been proposed as a therapeutic mechanism in triple-negative breast cancer, again with mixed results. Agents that target poly(ADP-ribose) polymerase and androgen receptors have also been proposed in these patients or subsets of them, and ongoing trials should result in definitive guidance with respect to the value of these agents in triple-negative disease. Triple-negative breast cancer is clearly a distinct clinical subtype, from the perspective of both ER and HER-2 expression, but further subclassification is needed. At present, there is not a clear, proven effective single agent that targets a defining vulnerability in triple-negative breast cancer. This article will review the clinical problem of triple-negative disease, potential prognostic factors, demonstrated efficacy of currently available therapeutic options, and new potential therapies.
Collapse
Affiliation(s)
- Clifford A Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, New York, USA.
| | | |
Collapse
|
38
|
Munagala R, Aqil F, Gupta RC. Promising molecular targeted therapies in breast cancer. Indian J Pharmacol 2011; 43:236-45. [PMID: 21713084 PMCID: PMC3113372 DOI: 10.4103/0253-7613.81497] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 02/23/2011] [Indexed: 12/27/2022] Open
Abstract
In recent years, there has been a significant improvement in the understanding of molecular events and critical pathways involved in breast cancer. This has led to the identification of novel targets and development of anticancer therapies referred to as targeted therapy. Targeted therapy has high specificity for the molecules involved in key molecular events that are responsible for cancer phenotype such as cell growth, survival, migration, invasion, metastasis, apoptosis, cell-cycle progression, and angiogenesis. Targeted agents that have been approved for breast cancer include trastuzumab and lapatinib, directed against human epidermal growth factor receptor 2 (HER2) and bevacizumab, directed against vascular endothelial growth factor (VEGF). Several other targeted agents currently under evaluation in preclinical and clinical trials include inhibitors of epidermal growth factor receptor (EGFR), dual EGFR and HER2 inhibitors, VEGF/VEGFR inhibitors, and agents that interfere with crucial signaling pathways such as PI3K/AKT/mTOR and RAS/MEK/ERK; agents against other tyrosine kinases such as Src, insulin-like growth factor (IGF)/IGF-receptor (IGFR); agents that promote apoptosis such as Poly ADP ribose polymerase inhibitors; agents that target invasion and metastasis such as matrix metalloproteinases inhibitors and others. In this review, we highlight the most promising targeted agents and their combination with mainstream chemotherapeutic drugs in clinical trials.
Collapse
Affiliation(s)
- Radha Munagala
- James Graham Brown Cancer Center, University of Louisville, Louisville
| | - Farrukh Aqil
- James Graham Brown Cancer Center, University of Louisville, Louisville
| | - Ramesh C. Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville
- Department of Pharmacology and Toxicology, University of Louisville, Louisville
| |
Collapse
|
39
|
Toft DJ, Cryns VL. Minireview: Basal-like breast cancer: from molecular profiles to targeted therapies. Mol Endocrinol 2011; 25:199-211. [PMID: 20861225 PMCID: PMC3035993 DOI: 10.1210/me.2010-0164] [Citation(s) in RCA: 118] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Accepted: 08/05/2010] [Indexed: 12/11/2022] Open
Abstract
The classification of breast cancer into molecular subtypes with distinctive gene expression signatures that predict treatment response and prognosis has ushered in a new era of personalized medicine for this remarkably heterogeneous and deadly disease. Basal-like breast cancer (BLBC) is a particularly aggressive molecular subtype defined by a robust cluster of genes expressed by epithelial cells in the basal or outer layer of the adult mammary gland. BLBC is a major clinical challenge because these tumors are prevalent in young woman, often relapsing rapidly. Additionally, most (but not all) basal-like tumors lack expression of steroid hormone receptors (estrogen receptor and progesterone receptor) and human epidermal growth factor receptor 2, limiting targeted therapeutic options for these predominantly triple-negative breast cancers. This minireview will focus on new insights into the molecular etiology of these poor-prognosis tumors that underlie their intrinsic genomic instability, deregulated cell proliferation and apoptosis, and invasive tumor biology. We will also review ongoing efforts to translate these fundamental insights into improved therapies for women with BLBC.
Collapse
Affiliation(s)
- Daniel J Toft
- Division of Endocrinology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 6061, USA
| | | |
Collapse
|
40
|
Subramaniam V, Ace O, Prud'homme GJ, Jothy S. Tranilast treatment decreases cell growth, migration and inhibits colony formation of human breast cancer cells. Exp Mol Pathol 2010; 90:116-22. [PMID: 21040720 DOI: 10.1016/j.yexmp.2010.10.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 10/21/2010] [Indexed: 10/18/2022]
Abstract
In the treatment of breast cancer, although a wide of choice of drugs and treatment modalities are available, drug resistance or drug toxicity poses a considerable challenge. Tranilast is a well tolerated drug used in the treatment of allergic disorders. Previous works in various models have shown that tranilast has the potential to be used as an anti-cancer drug. Hence, in this study using human breast cancer cell lines BT-474 and MDA-MB-231, we studied the effect of tranilast on cell growth, migration and ability to prevent colony formation in vitro, properties that are relevant to a possible therapeutic effect in breast cancer. We found that tranilast inhibits the growth of both breast cancer cell lines. In the cell migration experiments, the tumor cells exhibit significantly slower wound closure after tranilast treatment, as well as reduced migration using an insert system. Downregulation of MRTF-A, a global cytoskeleton regulator was observed after tranilast treatment. Additionally, tranilast treatment increased levels of cleaved PARP in both cell lines tested indicating a stimulation of apoptosis. A significant reduction in colony size and number was observed in soft agar clonogenic assays in both cell lines after tranilast treatment. BT-474 cells were more responsive to tranilast treatment compared to MDA-MB-231 cells, suggesting a difference in modes of action, or sensitivity, possibly related to their different receptor status. Based on these changes in cancer cell lines, we conclude that tranilast exerts effects that set a rationale for future preclinical studies in animal models of breast cancer.
Collapse
Affiliation(s)
- Venkateswaran Subramaniam
- Department of Laboratory Medicine and Keenan Research Centre of the Li Ka Shing Knowledge Institute, St Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
41
|
Saxena R, Dwivedi A. ErbB family receptor inhibitors as therapeutic agents in breast cancer: Current status and future clinical perspective. Med Res Rev 2010; 32:166-215. [DOI: 10.1002/med.20209] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
42
|
Abstract
Although the introduction of novel therapies and drug combinations has improved the prognosis of metastatic breast cancer, the disease remains incurable. Increased knowledge of the biology and the molecular alterations in breast cancer has facilitated the design of targeted therapies. These agents include receptor and nonreceptor tyrosine kinase inhibitors (epidermal growth factor receptor family), intracellular signaling pathways (phosphatidylinositol-3-kinase, AKT, mammalian target of rapamycin) angiogenesis inhibitors and agents that interfere with DNA repair (poly(ADP-ribose) polymerase inhibitors). In the present review, we present the most promising studies of these new targeted therapies and novel combinations of targeted therapies with cytotoxic agents.
Collapse
Affiliation(s)
- Ricardo H Alvarez
- Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1155 Herman P, Pressler, CPB5.3458, Houston, TX 77030-3721, USA.
| |
Collapse
|
43
|
Boér K, Láng I, Llombart-Cussac A, Andreasson I, Vivanco GL, Sanders N, Pover GM, Murray E. Vandetanib with docetaxel as second-line treatment for advanced breast cancer: a double-blind, placebo-controlled, randomized Phase II study. Invest New Drugs 2010; 30:681-7. [PMID: 20830502 DOI: 10.1007/s10637-010-9538-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 09/01/2010] [Indexed: 11/26/2022]
Abstract
PURPOSE The aim of this Phase II study was to assess the efficacy and safety of vandetanib in combination with docetaxel in patients with pretreated advanced breast cancer. METHODS The primary study objective was to compare the number of progression events in patients receiving once-daily oral vandetanib (100 mg) in combination with docetaxel (100 mg/m(2) iv every 21 days) versus placebo plus docetaxel. Sixty-four patients were randomized to receive study treatment (n = 35, vandetanib; n = 29, placebo). RESULTS A slightly greater number of patients had experienced a progression event by the data cut-off in the vandetanib group (24 [69%]) compared with the placebo group (18 [62%]); HR = 1.19, two-sided 80% CI: 0.79-1.81; two-sided P = 0.59), suggesting that the addition of vandetanib to docetaxel did not affect the risk of disease progression compared with placebo plus docetaxel. The safety and tolerability profile of the combination therapy reflected those of both drugs as monotherapy agents. CONCLUSIONS In patients with advanced breast cancer, vandetanib plus docetaxel was generally well tolerated. Clinical benefit was not different to that observed with placebo plus docetaxel. However, due to the small patient number it was not possible to yield robust results, further research is required to identify predictive factors for patient selection.
Collapse
Affiliation(s)
- Katalin Boér
- Medical Oncology Department, St John Hospital, Bécsi út 132, 1032 Budapest, Hungary.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Greenberg S, Rugo HS. Challenging Clinical Scenarios: Treatment of Patients With Triple-Negative or Basal-Like Metastatic Breast Cancer. Clin Breast Cancer 2010; 10 Suppl 2:S20-9. [DOI: 10.3816/cbc.2010.s.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Bauer JA, Ye F, Marshall CB, Lehmann BD, Pendleton CS, Shyr Y, Arteaga CL, Pietenpol JA. RNA interference (RNAi) screening approach identifies agents that enhance paclitaxel activity in breast cancer cells. Breast Cancer Res 2010; 12:R41. [PMID: 20576088 PMCID: PMC2917036 DOI: 10.1186/bcr2595] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Revised: 06/04/2010] [Accepted: 06/24/2010] [Indexed: 12/31/2022] Open
Abstract
Introduction Paclitaxel is a widely used drug in the treatment of patients with locally advanced and metastatic breast cancer. However, only a small portion of patients have a complete response to paclitaxel-based chemotherapy, and many patients are resistant. Strategies that increase sensitivity and limit resistance to paclitaxel would be of clinical use, especially for patients with triple-negative breast cancer (TNBC). Methods We generated a gene set from overlay of the druggable genome and a collection of genomically deregulated gene transcripts in breast cancer. We used loss-of-function RNA interference (RNAi) to identify gene products in this set that, when targeted, increase paclitaxel sensitivity. Pharmacological agents that targeted the top scoring hits/genes from our RNAi screens were used in combination with paclitaxel, and the effects on the growth of various breast cancer cell lines were determined. Results RNAi screens performed herein were validated by identification of genes in pathways that, when previously targeted, enhanced paclitaxel sensitivity in the pre-clinical and clinical settings. When chemical inhibitors, CCT007093 and mithramycin, against two top hits in our screen, PPMID and SP1, respectively, were used in combination with paclitaxel, we observed synergistic growth inhibition in both 2D and 3D breast cancer cell cultures. The transforming growth factor beta (TGFβ) receptor inhibitor, LY2109761, that targets the signaling pathway of another top scoring hit, TGFβ1, was synergistic with paclitaxel when used in combination on select breast cancer cell lines grown in 3D culture. We also determined the relative paclitaxel sensitivity of 22 TNBC cell lines and identified 18 drug-sensitive and four drug-resistant cell lines. Of significance, we found that both CCT007093 and mithramycin, when used in combination with paclitaxel, resulted in synergistic inhibition of the four paclitaxel-resistant TNBC cell lines. Conclusions RNAi screening can identify druggable targets and novel drug combinations that can sensitize breast cancer cells to paclitaxel. This genomic-based approach can be applied to a multitude of tumor-derived cell lines and drug treatments to generate requisite pre-clinical data for new drug combination therapies to pursue in clinical investigations.
Collapse
Affiliation(s)
- Joshua A Bauer
- Department of Biochemistry, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, 2200 Pierce Avenue, Nashville, TN 37232, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Alvarez RH, Valero V, Hortobagyi GN. Emerging targeted therapies for breast cancer. J Clin Oncol 2010; 28:3366-79. [PMID: 20530283 DOI: 10.1200/jco.2009.25.4011] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Increased understanding of the molecular events involved in cancer development has led to the identification of a large number of novel targets and, in parallel, to the development of multiple approaches to anticancer therapy. Targeted therapy focuses on specific molecules in the malignant cell signal transduction machinery, including crucial molecules involved in cell invasion, metastasis, apoptosis, cell-cycle control, and tumor-related angiogenesis. In breast cancer, two new targeted agents have recently been approved: lapatinib, directed against the human epidermal growth factor receptor 2 (HER2); and bevacizumab, directed against vascular endothelial growth factor (VEGF). Multiple other targeted agents are under evaluation in clinical trials, including inhibitors of the epidermal growth factor receptor (EGFR), dual EGFR and HER2 inhibitors, other VEGF or VEGF-receptor inhibitors, and agents that alter crucial signaling pathways, such as RAS/MEK/ERK; phosphatidylinositol-3-kinase/Akt/ mammalian target of rapamycin; insulin-like growth factor/insulin-like growth factor receptor; poly (ADP-ribose) polymerase 1; and others. In this review, we present the most promising studies of these new targeted therapies and novel combinations of targeted therapies with traditional cytotoxic agents.
Collapse
Affiliation(s)
- Ricardo H Alvarez
- The University of Texas M D Anderson Cancer Center, Department of Breast Medical Oncology, Houston, TX 77030-3721, USA.
| | | | | |
Collapse
|
47
|
Choo JR, Nielsen TO. Biomarkers for Basal-like Breast Cancer. Cancers (Basel) 2010; 2:1040-65. [PMID: 24281106 PMCID: PMC3835118 DOI: 10.3390/cancers2021040] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2010] [Revised: 05/11/2010] [Accepted: 05/19/2010] [Indexed: 12/24/2022] Open
Abstract
Initially recognized through microarray-based gene expression profiling, basal-like breast cancer, for which we lack effective targeted therapies, is an aggressive form of carcinoma with a predilection for younger women. With some success, immunohistochemical studies have attempted to reproduce the expression profile classification of breast cancer through identification of subtype-specific biomarkers. This review aims to present an in depth summary and analysis of the current status of basal-like breast cancer biomarker research. While a number of biomarkers show promise for future clinical application, the next logical step is a comprehensive investigation of all biomarkers against a gene expression profile gold standard for breast cancer subtype assignment.
Collapse
Affiliation(s)
- Jennifer R Choo
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada.
| | | |
Collapse
|
48
|
Epidermal growth factor receptor in triple-negative and basal-like breast cancer: promising clinical target or only a marker? Cancer J 2010; 16:23-32. [PMID: 20164687 DOI: 10.1097/ppo.0b013e3181d24fc1] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Triple-negative breast cancers represent a subset of breast cancers with a particularly aggressive phenotype and poor clinical outcomes. Recent molecular profiling of these tumors has revealed a high frequency of epidermal growth factor receptor (EGFR) dysregulation, among other abnormalities. EGFR status correlates negatively with survival in patients with triple-negative breast cancers, and thus focus has turned on this receptor as a potential clinical target. Two classes of EGFR inhibitors are currently in clinical use: the monoclonal antibodies and the small molecule tyrosine kinase inhibitors. Trials of these drugs in breast cancer, however, have been largely disappointing. It remains to be seen whether advances in our understanding of the mechanisms of EGFR dysregulation and effects of multiple compensatory pathways in breast cancer, coupled with improved targeting to appropriate patient populations, will yield meaningful improvements in clinical outcomes.
Collapse
|
49
|
Campos SM, Berlin ST, Parker LM, Chen WY, Bunnell CA, Atkinson T, Lee J, Matulonis U, Hirsch MS, Harris L, Krasner CN. Phase I trial of liposomal doxorubicin and ZD1839 in patients with refractory gynecological malignancies or metastatic breast cancer. Int J Clin Oncol 2010; 15:390-8. [PMID: 20405155 DOI: 10.1007/s10147-010-0073-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 03/08/2010] [Indexed: 11/29/2022]
Abstract
BACKGROUND Pegylated liposomal doxorubicin has activity in both breast and ovarian cancer. Preclinical data noted that ZD1839 acts synergistically with chemotherapy. Given the lack of cross-resistance between these two agents, a phase I trial was initiated examining the safety and efficacy of the combination of liposomal doxorubicin and ZD1839 in patients with recurrent gynecologic or metastatic breast cancer. METHODS Dose-limiting toxicity (DLT) was defined within the first two cycles of treatment. Escalating doses of liposomal doxorubicin were administered every 4 weeks with ZD1839. Pharmacokinetic analysis and correlative studies were performed. RESULTS Thirty-five patients were enrolled in this study: six in each cohort. One DLT (febrile neutropenia) was observed in cohort 2. Dose level 3 was determined to be the maximum tolerated dose (MTD), and an additional ten patients were accrued. Serious adverse events (SAEs) included one patient with mental status changes believed secondary to disease progression and two central nervous system (CNS) bleeds believed to be unrelated to the combination of study agents. Toxicities were generally mild except for skin and gastrointestinal toxicity. No cardiac toxicity was observed. The best response to therapy included four partial responses and 20 patients with stable disease. CONCLUSIONS Liposomal doxorubicin with ZD1839 is an active regimen but is associated with increased skin toxicity in patients with advanced breast and gynecologic cancer.
Collapse
Affiliation(s)
- Susana M Campos
- Department of Gynecology Oncology, Dana Farber Cancer Institute, 44 Binney Street, Boston, MA, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Carter CA, Kelly RJ, Giaccone G. Small-molecule inhibitors of the human epidermal receptor family. Expert Opin Investig Drugs 2010; 18:1829-42. [PMID: 19938898 DOI: 10.1517/13543780903373343] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Small molecule inhibitors of human epidermal receptors (HER) have become an integral part of the armamentarium available to the medical oncologist in the treatment of solid tumor malignancies. At present, there are two small-molecule inhibitors (erlotinib and lapatinib) approved by the FDA in the USA, and a third inhibitor, gefitinib, is approved in other countries. OBJECTIVE To summarize the current standards of care for these new agents in solid tumors, and to discuss ongoing clinical trials; to review the known mechanisms of action of these inhibitors as well as to discuss both the known predictive markers for response and likely mechanisms of resistance. METHODS We reviewed key presentations and recent publications on small-molecule inhibitors targeting the HER family in solid tumors. CONCLUSIONS Recent data have highlighted the importance of mutations and amplifications of receptors within the HER family. Amplification of HER2 often translates into responses in anti-HER2 therapy. Mutations either enhance sensitivity or confer resistance to small-molecule inhibitors. Other mechanisms of resistance are being elucidated which should lead to the ability to predict both responses and resistance to HER family inhibitors and should translate into improvements in patient care.
Collapse
Affiliation(s)
- Corey A Carter
- National Cancer Institute, NIH, Bethesda Naval Hospital, Building 10, Room 13N222, Bethesda, MD 20892, USA
| | | | | |
Collapse
|