51
|
Oualla K, El-Zawahry HM, Arun B, Reuben JM, Woodward WA, Gamal El-Din H, Lim B, Mellas N, Ueno NT, Fouad TM. Novel therapeutic strategies in the treatment of triple-negative breast cancer. Ther Adv Med Oncol 2017; 9:493-511. [PMID: 28717401 PMCID: PMC5502951 DOI: 10.1177/1758834017711380] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous subtype of breast cancer that is defined by negative estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) status. Treating patients with TNBC remains clinically challenging, as patients are not candidates for endocrine or HER2-directed therapy. As a result, chemotherapy with traditional agents such as anthracyclines and taxanes remains the only available option with moderate success. Recent discoveries have revealed that TNBC is a heterogeneous disease at the clinical, histological and molecular levels. The use of biomarkers to identify distinct subsets of TNBC that derive the greatest benefit from presently approved as well as novel therapeutics has become the main focus of current research. The aim of this review is to explore the clinical and biological complexity of TNBC as well as identify novel therapeutic options that target the various molecular subsets of TNBC.
Collapse
Affiliation(s)
- Karima Oualla
- Medical Oncology Department, Hassan II University Hospital, Fes, Morocco
| | - Heba M. El-Zawahry
- Department of Medical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Banu Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M. Reuben
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wendy A. Woodward
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heba Gamal El-Din
- Department of Surgical Oncology, The National Cancer Institute, Cairo University, Cairo, Egypt
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Nawfel Mellas
- Medical Oncology Department, Hassan II University Hospital, Fes, Morocco
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Tamer M. Fouad
- Department of Medical Oncology, The National Cancer Institute, Cairo University, Kasr El-Aini Road, Cairo, 11796, Egypt
| |
Collapse
|
52
|
Masuda M, Yamada T. Signaling pathway profiling using reverse-phase protein array and its clinical applications. Expert Rev Proteomics 2017. [PMID: 28621158 DOI: 10.1080/14789450.2017.1344101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Increased accessibility to next-generation sequencing within the last decade has led to a paradigm shift in cancer treatment from one-size-fits-all medicine to precision medicine providing therapeutic strategies tailored to the requirements of individual patients. However, the effect of even the most successful agent yet tested is only transient, and durable efficacy has yet to be achieved. Genome- and transcriptome-based approaches cannot fully predict the diversity of protein expression patterns or post-translational modifications that directly contribute to cancer pathogenesis and physiology. This underscores the need for concordant proteomic analysis in the next stage of precision medicine. Areas covered: This review begins with an overview of the recent advances and trends in precision medicine that currently rely on genomics, and highlights the utility of antibody-based reverse-phase protein array (RPPA) technology as a proteomic tool in this context. Expert commentary: RPPA is well suited for pharmacodynamics analysis in view of its ability to precisely map signaling status using limited amounts of clinical samples. In addition, the cost-effectiveness and rapid turn-around time of the RPPA platform offer a substantial advantage over existing molecular profiling technologies in clinical settings.
Collapse
Affiliation(s)
- Mari Masuda
- a Division of Chemotherapy and Clinical Research , National Cancer Center Research Institute , Tokyo , Japan
| | - Tesshi Yamada
- a Division of Chemotherapy and Clinical Research , National Cancer Center Research Institute , Tokyo , Japan
| |
Collapse
|
53
|
Li X, Oprea-Ilies GM, Krishnamurti U. New Developments in Breast Cancer and Their Impact on Daily Practice in Pathology. Arch Pathol Lab Med 2017; 141:490-498. [DOI: 10.5858/arpa.2016-0288-sa] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Advances in research have transformed our understanding of breast cancers and have altered the daily practice of pathology. Theranostic evaluations performed by pathologists are now critical in triaging the patients into appropriate treatment groups, as are new guidelines that were recently established for the evaluation of HER2/neu gene amplification. Emerging molecular classifications of breast cancers bring novel perspectives to the assessment of individual cases, and opportunities for better treatments. Molecular studies have particularly shed light on distinct biological subsets of triple-negative breast cancers, for which new targeted therapies are being developed. The prognostic and therapeutic utility of new histopathologic parameters, such as tumor-infiltrating lymphocytes, are also being elucidated, and new protocols have been devised for the pathologic evaluation of breast specimens that have undergone neoadjuvant treatment. Novel clinical practices, such as radioactive seed localization, also affect the way breast specimens are processed and evaluated. In this brief review, we highlight the developments that are most relevant to pathology and are changing or could potentially impact our daily practice.
Collapse
Affiliation(s)
| | | | - Uma Krishnamurti
- From the Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Gerogia
| |
Collapse
|
54
|
Neoadjuvant systemic therapy in breast cancer: Challenges and uncertainties. Eur J Obstet Gynecol Reprod Biol 2016; 210:144-156. [PMID: 28039758 DOI: 10.1016/j.ejogrb.2016.12.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 11/20/2016] [Accepted: 12/13/2016] [Indexed: 12/29/2022]
Abstract
The management of locally advanced breast cancer (LABC) remains a major clinical issue, despite progress achieved in diagnosis and therapy. Preoperative or neoadjuvant therapy has gained interest since breast cancer has been regarded as a systemic disease. Comparing adjuvant versus neoadjuvant treatment, the neoadjuvant approach offers the advantage of downstaging the disease and testing the efficacy of therapy administered to patients. A large number of clinical trials have attempted to define the optimal neoadjuvant treatment, but little attention has been paid to the sequence of chemotherapy. Moreover, the integration of antibodies against Human Epidermal Receptor-2 (HER-2) and other biological therapies that may improve the long-term control of breast cancer patients, have a special clinical interest. In this review, we will discuss these topics attempting to answer the questions why, when and which regimen to use for patients with LABC. Especially, the introduction of the platina derivatives in neoadjuvant trials with their exceptional high pathological complete response rates are challenging to rethink the optimal treatment options in early and locally advanced breast cancer.
Collapse
|
55
|
Chamberlin MD, Bernhardt EB, Miller TW. Clinical Implementation of Novel Targeted Therapeutics in Advanced Breast Cancer. J Cell Biochem 2016; 117:2454-63. [PMID: 27146558 PMCID: PMC6010350 DOI: 10.1002/jcb.25590] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/19/2022]
Abstract
The majority of advanced breast cancers have genetic alterations that are potentially targetable with drugs. Through initiatives such as The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), data can be mined to provide context for next-generation sequencing (NGS) results in the landscape of advanced breast cancer. Therapies for targets other than estrogen receptor alpha (ER) and HER2, such as cyclin-dependent kinases CDK4 and CDK6, were recently approved based on efficacy in patient subpopulations, but no predictive biomarkers have been found, leaving clinicians to continue a trial-and-error approach with each patient. Next-generation sequencing identifies potentially actionable alterations in genes thought to be drivers in the cancerous process including phosphatidylinositol 3-kinase (PI3K), AKT, fibroblast growth factor receptors (FGFRs), and mutant HER2. Epigenetically directed and immunologic therapies have also shown promise for the treatment of breast cancer via histone deacetylases (HDAC) 1 and 3, programmed T cell death 1 (PD-1), and programmed T cell death ligand 1 (PD-L1). Identifying biomarkers to predict primary resistance in breast cancer will ultimately affect clinical decisions regarding adjuvant therapy in the first-line setting. However, the bulk of medical decision-making is currently made in the secondary resistance setting. Herein, we review the clinical potential of PI3K, AKT, FGFRs, mutant HER2, HDAC1/3, PD-1, and PD-L1 as therapeutic targets in breast cancer, focusing on the rationale for therapeutic development and the status of clinical testing. J. Cell. Biochem. 117: 2454-2463, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mary D Chamberlin
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.
- Department of Hematology-Oncology, One Medical Center Dr., Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire.
- Comprehensive Breast Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.
| | - Erica B Bernhardt
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Todd W Miller
- Comprehensive Breast Program, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| |
Collapse
|
56
|
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an aggressive breast cancer subtype. However, there have been limited data to evaluate the trend of TNBC research. This study aims to investigate the trend of TNBC research and compare the contribution of research from different regions, organizations, and authors. METHODS TNBC-related publications from 2007 to 2015 were retrieved from the Web of Science database. Excel 2013 (Redmond, Washington, USA), GraphPad Prism 5 (GraphPad Prism Software Inc., San Diego, CA), and VOSviewer (Leiden University, Leiden, Netherlands) software were used to analyze the trend of TNBC research. This article does not contain any studies with human participants or animals performed by any of the authors. RESULTS A total of 1695 papers were identified and were cited 34,078 times with a time limit of May 27, 2016. The United States accounted for 43.10% of the articles, 57.59% of the citations, and the highest H-index (64). China ranked second in total number of articles, but seventh in citation frequency (1998) and ninth in H-index (21). The journal Breast Cancer Research and Treatment had the highest number of publications. The author, Narod SA, has published the most papers in this field (30). The keyword "receptor" was mentioned the most, 1489 times, and the word "myeloid cell leukemia-1 (MCL-1)" was the latest hot spot by 2015. CONCLUSION Literature growth related to TNBC is expanding rapidly in recent years. The quality of the articles from China still requires improvement. Newest progress of the TNBC research may be released by the journal Breast Cancer Research and Treatment first. Narod SA, Gonzalez-Angulo AM, and Hortobagyi GN may be good candidates for collaborative research in this field. MCL-1 is an emerging topic that should be closely observed.
Collapse
Affiliation(s)
- Yiran Wang
- Graduate Management Unit
- Department of Oncology
| | - Xiao Zhai
- Graduate Management Unit
- Department of Orthopedics, Changhai Hospital Affiliated to the Second Military Medical University
| | - Chuan Liu
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | |
Collapse
|
57
|
Triple-negative breast cancer: advancements in characterization and treatment approach. Curr Opin Obstet Gynecol 2016; 28:59-69. [PMID: 26694831 DOI: 10.1097/gco.0000000000000239] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW Triple-negative breast cancer (TNBC) comprises 15-20% of all breast cancer and is defined by the lack of estrogen and progesterone receptor expression and absence of human epidermal growth factor receptor 2 amplification. Compared with patients with hormone receptor positive or Her-2 positive breast cancer, patients with TNBC are more commonly young (age <50 years), African-American and have a higher incidence of BRCA1/2 mutations. The clinical course is frequently characterized by early relapse and poor overall survival. The TNBC phenotype is impervious to therapies commonly used in other breast cancer subtypes, including hormonal therapy and Her-2 receptor antagonism. Cytotoxic chemotherapy remains the only approved treatment. With its aggressive clinical course and paucity of effective treatment options, TNBC represents an unmet clinical need. This review will focus on updates of the biologic underpinnings of TNBC and the associated treatment advances. RECENT FINDINGS Numerous advancements have been made toward understanding the biologic framework of TNBC. Gene expression profiling has revealed six clinically relevant subsets of TNBC. Further study has demonstrated a portion of TNBC exhibits a strong immune gene signature. Lastly, it is now appreciated that a subgroup of sporadic TNBC shares biologic characteristics with BRCA1/2-mutated breast cancer, notably homologous repair deficiency. Recent studies focus on incorporation of platinum salts and new combinations of conventional chemotherapeutic agents. Targeted agents, including poly-ADP ribose polymerase inhibitors, antiangiogenic agents, phosphoinositide 3-kinase (PI3K) pathway inhibitors, and androgen antagonist are also being evaluated. Most recently, checkpoint inhibitors have demonstrated a modest degree of activity in a subset of TNBC. SUMMARY These discoveries are informing novel treatment paradigms and identification of correlative biomarkers in TNBC. Improved understanding of the biologic heterogeneity of TNBC is allowing for a more effective and individualized approach to treatment.
Collapse
|
58
|
Massihnia D, Galvano A, Fanale D, Perez A, Castiglia M, Incorvaia L, Listì A, Rizzo S, Cicero G, Bazan V, Castorina S, Russo A. Triple negative breast cancer: shedding light onto the role of pi3k/akt/mtor pathway. Oncotarget 2016; 7:60712-60722. [PMID: 27474173 PMCID: PMC5312414 DOI: 10.18632/oncotarget.10858] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/14/2016] [Indexed: 12/22/2022] Open
Abstract
Breast cancer is one of the most widespread carcinoma and one of the main causes of cancer-related death worldwide, especially in women aged between 35 and 75 years. Among the different subtypes, triple negative breast cancer (TNBC) is characterized by the total absence of the estrogen-receptor (ER) and progesteron-receptor (PR) expression as well as the lack of human epidermal growth factor receptor 2 (HER2) overexpression or gene amplification. These biological characteristics confer to TNBC a higher aggressiveness and relapse risk along with poorer prognosis compared to other subtypes. Indeed, 5-years survival rate is still low and almost all patients die, despite any adjuvant treatment which at moment represents the heading pharmacological approach. To date, several clinical trials have been designed to investigate the potential role of some molecular markers, such as VEGF, EGFR, Src and mTOR, for targeted treatments in TNBC. In fact, many inhibitors of the PI3K/AKT/mTOR pathway, frequently de-regulated in TNBC, are acquiring a growing interest and several inhibitors are in preclinical development or already in early phase clinical trials. In this Review, we investigated the role of the PI3K/AKT/mTOR pathway in TNBC patients, by summarizing the molecular features that led to the distinction of different histotypes of TNBC. Furthermore, we provided an overview of the inhibition mechanisms of the mTOR and PI3K/AKT signaling pathways, highlighting the importance of integrating biological and clinical data for the development of mTOR inhibitors in order to implement targeted therapies for TNBC patients.
Collapse
Affiliation(s)
- Daniela Massihnia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alessandro Perez
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Marta Castiglia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Angela Listì
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Sergio Rizzo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Giuseppe Cicero
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Sergio Castorina
- Fondazione Mediterranea “G.B. Morgagni”, Catania, Italy
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| |
Collapse
|
59
|
Quintela-Fandino M, Lluch A, Manso L, Calvo I, Cortes J, García-Saenz JA, Gil-Gil M, Martinez-Jánez N, Gonzalez-Martin A, Adrover E, de Andres R, Viñas G, Llombart-Cussac A, Alba E, Guerra J, Bermejo B, Zamora E, Moreno-Anton F, Pernas Simon S, Carrato A, Lopez-Alonso A, Escudero MJ, Campo R, Carrasco E, Palacios J, Mulero F, Colomer R. 18F-fluoromisonidazole PET and Activity of Neoadjuvant Nintedanib in Early HER2-Negative Breast Cancer: A Window-of-Opportunity Randomized Trial. Clin Cancer Res 2016; 23:1432-1441. [DOI: 10.1158/1078-0432.ccr-16-0738] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/12/2016] [Accepted: 07/19/2016] [Indexed: 11/16/2022]
|
60
|
Triple negative breast cancer: looking for the missing link between biology and treatments. Oncotarget 2016; 6:26560-74. [PMID: 26387133 PMCID: PMC4694936 DOI: 10.18632/oncotarget.5306] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 08/24/2015] [Indexed: 12/11/2022] Open
Abstract
The so called “Triple Negative Breast Cancer” (TNBC) represents approximately 15-20% of breast cancers. This acronym simply means that the tumour does not express oestrogen receptor (ER) and progesterone receptor (PR) and does not exhibit amplification of the human epidermal growth factor receptor 2 (HER2) gene. Despite this unambiguous definition, TNBCs are an heterogeneous group of tumours with just one common clinical feature: a distinctly aggressive nature with higher rates of relapse and shorter overall survival in the metastatic setting compared with other subtypes of breast cancer. Because of the absence of well-defined molecular targets, cytotoxic chemotherapy is currently the only treatment option for TNBC. In the last decades, the use of more aggressive chemotherapy has produced a clear improvement of the prognosis in women with TNBC, but this approach results in an unacceptable deterioration in the quality of life, also if some support therapies try to relieve patients from distress. In addition, there is the general belief that it is impossible to further improve the prognosis of TNBC patients with chemotherapy alone. In view of that, there is a feverish search for new “clever drugs” able both to rescue chemo-resistant, and to reduce the burden of chemotherapy in chemo-responsive TNBC patients. A major obstacle to identifying actionable targets in TNBC is the vast disease heterogeneity both inter-tumour and intra-tumour and years of study have failed to demonstrate a single unifying alteration that is targetable in TNBC. TNBC is considered the subtype that best benefits from the neoadjuvant model, since the strong correlation between pathological Complete Response and long-term Disease-Free-Survival in these patients. In this review, we discuss the recent discoveries that have furthered our understanding of TNBC, with a focus on the subtyping of TNBC. We also explore the implications of these discoveries for future treatments and highlight the need for a completely different type of clinical trials.
Collapse
|
61
|
Abstract
NEOADJUVANT CHEMOTHERAPY IN HER2-POSITIVE AND TRIPLE-NEGATIVE BREAST CANCER: Neoadjuvant chemotherapy is a standard option in the management of operable breast cancer, as effective as adjuvant chemotherapy in term of survival and with the potential to increase the rate of breast conservation. In HER2+ and triple-negative breast cancers, neoadjuvant chemotherapy is associated with a high probability of pathological complete response, which strongly predicts survival outcome. In HER2+ breast cancer, trastuzumab, in combination with neoadjuvant chemotherapy, mostly anthracyclines and taxane-based, demonstrated a significant increase in pathological complete response rate. Recently, dual HER2 blockade strategies (lapatinib-trastuzumab or pertuzumab-trastuzumab) demonstrated a significant improvement in terms of pathological complete response over trastuzumab. In triple-negative breast cancer, recent data indicate that incorporating platinum compounds to neoadjuvant chemotherapy also significantly improves this parameter. Yet, in both subtypes, whether these substantial improvements may lead to significant benefits in terms of survival and breast conserving surgery remains to be demonstrated.
Collapse
Affiliation(s)
- Anthony Gonçalves
- Département d'oncologie médicale, Institut Paoli-Calmettes, Marseille, 13009; Centre de recherche en cancérologie de Marseille (Institut Paoli-Calmettes, Inserm 1068, Cnrs 7258, Aix-Marseille Université), Marseille, 13009; Aix-Marseille Université, Marseille, 13006.
| |
Collapse
|
62
|
Študentová H, Vitásková D, Melichar B. Safety of mTOR inhibitors in breast cancer. Expert Opin Drug Saf 2016; 15:1075-85. [DOI: 10.1080/14740338.2016.1192604] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Hana Študentová
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| | - Denisa Vitásková
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| | - Bohuslav Melichar
- Department of Oncology, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
- Institute of Molecular and Translational Medicine, Palacký University Medical School & Teaching Hospital, Olomouc, Czech Republic
| |
Collapse
|
63
|
Zhang X, Ran YG, Wang KJ. Risk of mTOR inhibitors induced severe pneumonitis in cancer patients: a meta-analysis of randomized controlled trials. Future Oncol 2016; 12:1529-39. [PMID: 27067269 DOI: 10.2217/fon-2016-0020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background: A meta-analysis of randomized controlled trials was performed to determine the overall risk of noninfectious severe pneumonitis associated with mTOR inhibitors (mTORi) in cancer patients. Materials & methods: PubMed, EMBASE and oncology conference proceedings were searched for relevant studies. Results: A total of 8377 patients from 16 randomized controlled trials were included. The incidence of severe pneumonitis associated with mTORi was 1.7% (95% CI: 1.1–2.5%). The use of mTORi significantly increased the risk of severe pneumonitis compared with controls (odds ratio: 3.36; 95% CI: 2.20–5.12). The analysis was stratified for drug types, tumor types, controlled therapy and mTORi-based regimens, but no significant differences in odds ratios were observed. Conclusion: mTORi significantly increase the risk of severe pneumonitis in cancer patients.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Yu-ge Ran
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding 071000, China
| | - Kun-jie Wang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, China
| |
Collapse
|
64
|
Steelman LS, Martelli AM, Cocco L, Libra M, Nicoletti F, Abrams SL, McCubrey JA. The therapeutic potential of mTOR inhibitors in breast cancer. Br J Clin Pharmacol 2016; 82:1189-1212. [PMID: 27059645 DOI: 10.1111/bcp.12958] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/29/2016] [Accepted: 03/31/2016] [Indexed: 12/16/2022] Open
Abstract
Rapamycin and modified rapamycins (rapalogs) have been used to prevent allograft rejection after organ transplant for over 15 years. The mechanistic target of rapamycin (mTOR) has been determined to be a key component of the mTORC1 complex which consists of the serine/threonine kinase TOR and at least five other proteins which are involved in regulating its activity. Some of the best characterized substrates of mTORC1 are proteins which are key kinases involved in the regulation of cell growth (e.g., p70S6K) and protein translation (e.g., 4E-BP1). These proteins may in some cases serve as indicators to sensitivity to rapamycin-related therapies. Dysregulation of mTORC1 activity frequently occurs due to mutations at, or amplifications of, upstream growth factor receptors (e.g., human epidermal growth factor receptor-2, HER2) as well as kinases (e.g., PI3K) and phosphatases (e.g., PTEN) critical in the regulation of cell growth. More recently, it has been shown that certain rapalogs may enhance the effectiveness of hormonal-based therapies for breast cancer patients who have become resistant to endocrine therapy. The combined treatment of certain rapalogs (e.g., everolimus) and aromatase inhibitors (e.g., exemestane) has been approved by the United States Food and Drug Administration (US FDA) and other drug regulatory agencies to treat estrogen receptor positive (ER+) breast cancer patients who have become resistant to hormonal-based therapies and have progressed. This review will summarize recent basic and clinical research in the area and evaluate potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Linda S Steelman
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - Alberto M Martelli
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Dipartimento di Scienze Biomediche e Neuromotorie, Università di Bologna, Bologna, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, University of Catania, Catania, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology & Functional Genomics, Section of Pathology & Oncology, University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine at East Carolina University, Greenville, NC, USA.
| |
Collapse
|
65
|
Taxane acute pain syndrome (TAPS) in patients receiving taxane-based chemotherapy for breast cancer—a systematic review. Support Care Cancer 2016; 24:3633-50. [DOI: 10.1007/s00520-016-3256-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/26/2016] [Indexed: 12/19/2022]
|
66
|
Lee JJ, Loh K, Yap YS. PI3K/Akt/mTOR inhibitors in breast cancer. Cancer Biol Med 2015; 12:342-54. [PMID: 26779371 PMCID: PMC4706528 DOI: 10.7497/j.issn.2095-3941.2015.0089] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022] Open
Abstract
Activation of the phosphoinositide 3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is common in breast cancer. There is preclinical data to support inhibition of the pathway, and phase I to III trials involving inhibitors of the pathway have been or are being conducted in solid tumors and breast cancer. Everolimus, an mTOR inhibitor, is currently approved for the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. In this review, we summarise the efficacy and toxicity findings from the randomised clinical trials, with simplified guidelines on the management of potential adverse effects. Education of healthcare professionals and patients is critical for safety and compliance. While there is some clinical evidence of activity of mTOR inhibition in HR-positive and HER2-positive breast cancers, the benefits may be more pronounced in selected subsets rather than in the overall population. Further development of predictive biomarkers will be useful in the selection of patients who will benefit from inhibition of the PI3K/Akt/mTOR (PAM) pathway.
Collapse
Affiliation(s)
- Joycelyn Jx Lee
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| | - Kiley Loh
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| | - Yoon-Sim Yap
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| |
Collapse
|
67
|
Nounou MI, ElAmrawy F, Ahmed N, Abdelraouf K, Goda S, Syed-Sha-Qhattal H. Breast Cancer: Conventional Diagnosis and Treatment Modalities and Recent Patents and Technologies. Breast Cancer (Auckl) 2015; 9:17-34. [PMID: 26462242 PMCID: PMC4589089 DOI: 10.4137/bcbcr.s29420] [Citation(s) in RCA: 119] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Breast cancer is the most prevalent cancer among women worldwide. However, increased survival is due to the dramatic advances in the screening methods, early diagnosis, and breakthroughs in treatments. Over the course of the last decade, many acquisitions have taken place in this critical field of research in the pharmaceutical industry. Advances in molecular biology and pharmacology aided in better understanding of breast cancer, enabling the design of smarter therapeutics able to target cancer and respond to its microenvironment efficiently. Patents and research papers investigating diagnosis and treatment strategies for breast cancer using novel technologies have been surveyed for the past 15 years. Various nanocarriers have been introduced to improve the therapeutic efficacy of anticancer drugs, including liposomes, polymeric micelles, quantum dots, nanoparticles, and dendrimers. This review provides an overview of breast cancer, conventional therapy, novel technologies in the management of breast cancer, and rational approaches for targeting breast cancer. HIGHLIGHTS Breast cancer is the most common cancer in women worldwide. However, survival rates vary widely, optimistically heading toward a positive trend. Increased survival is due to the drastic shift in the screening methods, early diagnosis, and breakthroughs in treatments.Different strategies of breast cancer classification and staging have evolved over the years. Intrinsic (molecular) subtyping is essential in clinical trials and well understanding of the disease.Many novel technologies are being developed to detect distant metastases and recurrent disease as well as to assess response to breast cancer management.Intensive research efforts are actively ongoing to take novel breast cancer therapeutics to potential clinical application.Most of the recent research papers and patents discuss one of the following strategies: the development of new drug entities that specifically target the breast tumor cells; tailor designing a novel carrier system that can multitask and multifunction as a drug carrier, targeting vehicle and even as a diagnostic tool, direct conjugation of a therapeutic drug moiety with a targeting moiety, diagnostic moiety or pharmacokinetics altering moiety; or the use of innovative nontraditional approaches such as genetic engineering, stem cells, or vaccinations.
Collapse
Affiliation(s)
- Mohamed I. Nounou
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Fatema ElAmrawy
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Nada Ahmed
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Kamilia Abdelraouf
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | | | | |
Collapse
|
68
|
Royce ME, Osman D. Everolimus in the Treatment of Metastatic Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2015; 9:73-9. [PMID: 26417203 PMCID: PMC4571987 DOI: 10.4137/bcbcr.s29268] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 06/14/2015] [Indexed: 02/03/2023]
Abstract
The discovery of the mammalian target of rapamycin (mTOR) molecular pathway has brought insight into its vital role in breast cancer pathogenesis. Several clinical trials have shown that the mTOR inhibitor everolimus could improve patient outcomes in several subtypes of breast cancer, including hormone receptor–positive, human epidermal growth factor receptor–negative metastatic disease that has progressed after prior endocrine therapy. This review summarizes findings from clinical trials that have demonstrated the benefit of everolimus in metastatic breast cancer and highlights some new research directions utilizing everolimus.
Collapse
Affiliation(s)
- Melanie E Royce
- Multidisciplinary Breast Cancer Clinic and Program, University of New Mexico Cancer Center, Albuquerque, NM, USA
| | - Diaa Osman
- Multidisciplinary Breast Cancer Clinic and Program, University of New Mexico Cancer Center, Albuquerque, NM, USA
| |
Collapse
|
69
|
Wang J, Zhang C, Chen K, Tang H, Tang J, Song C, Xie X. ERβ1 inversely correlates with PTEN/PI3K/AKT pathway and predicts a favorable prognosis in triple-negative breast cancer. Breast Cancer Res Treat 2015; 152:255-69. [PMID: 26070269 DOI: 10.1007/s10549-015-3467-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 06/09/2015] [Indexed: 02/06/2023]
Abstract
In contrast to the well-established role of estrogen receptor alpha (ERα) in breast cancer, the significance of estrogen receptor beta (ERβ) remains controversial, especially in triple-negative breast cancer (TNBC). We sought to investigate the clinical importance of wild-type ERβ (ERβ1) in TNBC based on a large population, and to explore the potential molecular pathways involved in. A total of 571 patients with invasive TNBC undergoing curative surgery were included in this study. Immunohistochemical staining for ERβ1, pAKT, PTEN, pERK, β-catenin, EGFR, p53, and E-cadherin was performed on tissue microarrays. Prognostic determinants for overall survival (OS) and disease-free survival (DFS), as well as the risk factors for distant metastasis-free survival (DMFS) and locoregional recurrence-free survival, were evaluated in univariate and multivariate analyses. Overexpression of ERβ1 was detected in 30.4% of tumor samples. Patients with ERβ1 tended to be postmenopausal, and less likely to develop lymphatic metastasis. Multivariate analysis demonstrated that ERβ1 predicted a better OS, DFS, and DMFS independently. Regarding other biomarkers, only pAKT was identified as an independent negative predictor for survival. Additionally, ERβ1 expression was inversely associated with pAKT and the loss of PTEN. Notably, further survival analysis according to status of ERβ1/pAKT indicated that ERβ1(+)/pAKT(-) predicted the most favorable prognosis for TNBC. On the contrary, ERβ1(-)/pAKT(+) was associated with the worst outcomes. In summary, our findings indicate that ERβ1 independently predicts a better prognosis for TNBC and potentially interacts with the PTEN/PI3K/pAKT pathway. The role of ERβ1-specific agonists combined with the inhibitors of pAKT merits further investigation.
Collapse
Affiliation(s)
- Jin Wang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, No. 651 Dongfeng East Road, Yuexiu District, Guangzhou, 510060, China,
| | | | | | | | | | | | | |
Collapse
|
70
|
Abstract
INTRODUCTION Triple negative breast cancer (TNBC) is a heterogeneous disease associated with a high risk of recurrence, and therapeutic options are currently limited to cytotoxic therapy. Germ-line mutations may occur in up to 20% of unselected patients with TNBC, which may serve as a biomarker identifying which patients may have tumors that are particularly sensitive to platinums and/or inhibitors of poly(ADP-ribose)polymerase. A substantial proportion of patients with TNBCs not associated with germ-line BRCA mutations may have tumors that are ‘BRCA-like’, rendering those individuals potential candidates for similar strategies. AREAS COVERED The purpose of this review is to highlight the current standard and experimental treatment strategies. EXPERT OPINION Recent research that has illuminated the molecular heterogeneity of the disease rationalizes its diverse biological behavior and differential response to chemotherapy. Modern technology platforms provide molecular signatures that can be mined for therapeatic interventions. Target pathways that are commonly dysregulated in cancer cells control cellular processes such as apoptosis, proliferation, angiogenesis, DNA repair, cell cycle progression, immune modulation and invasion, and metastasis. Novel trial design and re-defined endpoints as surrogates to clinical outcome have been introduced to expedite the development of breakthrough therapies to treat high-risk early-stage breast cancer.
Collapse
Affiliation(s)
- Eleni Andreopoulou
- Associate Professor of Medicine, Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, 1695 Eastchester Rd Bronx, NY 10461 USA
| | - Sarah J Schweber
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| | - Joseph A Sparano
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| | - Hayley M McDaid
- Montefiore Medical Center/Albert Einstein College of Medicine, Department of Medical Oncology, Bronx, NY, USA
| |
Collapse
|
71
|
O'Reilly EA, Gubbins L, Sharma S, Tully R, Guang MHZ, Weiner-Gorzel K, McCaffrey J, Harrison M, Furlong F, Kell M, McCann A. The fate of chemoresistance in triple negative breast cancer (TNBC). BBA CLINICAL 2015; 3:257-75. [PMID: 26676166 PMCID: PMC4661576 DOI: 10.1016/j.bbacli.2015.03.003] [Citation(s) in RCA: 272] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Treatment options for women presenting with triple negative breast cancer (TNBC) are limited due to the lack of a therapeutic target and as a result, are managed with standard chemotherapy such as paclitaxel (Taxol®). Following chemotherapy, the ideal tumour response is apoptotic cell death. Post-chemotherapy, cells can maintain viability by undergoing viable cellular responses such as cellular senescence, generating secretomes which can directly enhance the malignant phenotype. SCOPE OF REVIEW How tumour cells retain viability in response to chemotherapeutic engagement is discussed. In addition we discuss the implications of this retained tumour cell viability in the context of the development of recurrent and metastatic TNBC disease. Current adjuvant and neo-adjuvant treatments available and the novel potential therapies that are being researched are also reviewed. MAJOR CONCLUSIONS Cellular senescence and cytoprotective autophagy are potential mechanisms of chemoresistance in TNBC. These two non-apoptotic outcomes in response to chemotherapy are inextricably linked and are neglected outcomes of investigation in the chemotherapeutic arena. Cellular fate assessments may therefore have the potential to predict TNBC patient outcome. GENERAL SIGNIFICANCE Focusing on the fact that cancer cells can bypass the desired cellular apoptotic response to chemotherapy through cellular senescence and cytoprotective autophagy will highlight the importance of targeting non-apoptotic survival pathways to enhance chemotherapeutic efficacy.
Collapse
Affiliation(s)
- Elma A O'Reilly
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Luke Gubbins
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Shiva Sharma
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland ; Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Riona Tully
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Matthew Ho Zhing Guang
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - Karolina Weiner-Gorzel
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| | - John McCaffrey
- Department of Oncology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Michele Harrison
- Department of Pathology, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Fiona Furlong
- School of Pharmacy, Queens University Belfast, Belfast BT7 1NN, UK
| | - Malcolm Kell
- Department of Surgery, Mater Misericordiae Hospital, Dublin 7, Ireland
| | - Amanda McCann
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine and Medical Science (SMMS), Belfield, Dublin 4, Ireland
| |
Collapse
|
72
|
Patil A, Sherbet G. Therapeutic approach to the management of HER2-positive breast cancer metastatic to the brain. Cancer Lett 2015; 358:93-99. [DOI: 10.1016/j.canlet.2014.12.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 01/06/2023]
|
73
|
Gonzalez-Angulo AM, Krop I, Akcakanat A, Chen H, Liu S, Li Y, Culotta KS, Tarco E, Piha-Paul S, Moulder-Thompson S, Velez-Bravo V, Sahin AA, Doyle LA, Do KA, Winer EP, Mills GB, Kurzrock R, Meric-Bernstam F. SU2C phase Ib study of paclitaxel and MK-2206 in advanced solid tumors and metastatic breast cancer. J Natl Cancer Inst 2015; 107:dju493. [PMID: 25688104 DOI: 10.1093/jnci/dju493] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND There is preclinical synergism between taxanes and MK-2206. We aim to determine the maximum tolerated dose, safety, and activity of combining MK-2206 and paclitaxel in metastatic cancer. METHODS Patients received weekly doses of paclitaxel at 80mg/m2 on day 1, followed by MK-2206 orally on day 2 escalated at 90mg, 135mg, and 200mg. Treatment continued until progression, excessive toxicity, or patient request. Blood and tissue were collected for pharmacokinetic and pharmacodynamics markers. A cycle consisted of three weeks of therapy. Dose-limiting toxicity (DLT) was defined as unacceptable toxicity during the first cycle. All statistical tests were two-sided. RESULTS Twenty-two patients were treated, nine in dose escalation and 13 in dose expansion. Median age was 55 years. Median number of cycles was four. Dose escalation was completed with no DLT. CTCAE Grade 3 or higher adverse events were fatigue (n = 2), rash (n = 2), hyperglycemia (n = 1), and neutropenia (n = 7). Four patients in the expansion phase required MK-2206 dose reduction. Phase II recommended dose was established as paclitaxel 80mg/m2 weekly on day 1, and MK-2206 135mg weekly on day 2. Paclitaxel systemic exposure was similar in the presence or absence of MK-2206. Plasma MK-2206 concentrations were similar to data from previous phase I monotherapy. There was a statistically significant decrease in expression of pAKT S473 (P = .01) and pAKT T308 (P = .002) after therapy. PI3K/AKT/mTOR downregulation in tumor tissues and circulating markers did not correlate with tumor response or clinical benefit. There were five objective responses, and nine patients had stable disease. CONCLUSION MK-2206 was well tolerated with paclitaxel. Preliminary antitumor activity was documented.
Collapse
Affiliation(s)
- Ana M Gonzalez-Angulo
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Ian Krop
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Argun Akcakanat
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Huiqin Chen
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Shuying Liu
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Yisheng Li
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Kirk S Culotta
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Emily Tarco
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Sarina Piha-Paul
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Stacy Moulder-Thompson
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Vivianne Velez-Bravo
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Aysegul A Sahin
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Laurence A Doyle
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Kim-Anh Do
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Eric P Winer
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Gordon B Mills
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Razelle Kurzrock
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD)
| | - Funda Meric-Bernstam
- Departments of Breast Medical Oncology (AMGA, SL, SMT), Systems Biology (AMGA, GBM), Biostatistics (HC, YL, K-AD), Experimental Therapeutics (KSC), Investigational Cancer Therapeutics (AA, ET, SPP, VV-B, FMB), Pathology (AAS) and Surgical Oncology (FMB), The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Hematology-Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (IK, EPW); Division of Hematology-Oncology, University of California, San Diego, CA (RK); Cancer Therapy Evaluation Program, NIH/National Cancer Institute, Rockville, MD (LAD).
| |
Collapse
|
74
|
Tomao F, Papa A, Zaccarelli E, Rossi L, Caruso D, Minozzi M, Vici P, Frati L, Tomao S. Triple-negative breast cancer: new perspectives for targeted therapies. Onco Targets Ther 2015; 8:177-93. [PMID: 25653541 PMCID: PMC4303459 DOI: 10.2147/ott.s67673] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is a heterogeneous disease, encompassing a large number of entities showing different morphological features and having clinical behaviors. It has became apparent that this diversity may be justified by distinct patterns of genetic, epigenetic, and transcriptomic aberrations. The identification of gene-expression microarray-based characteristics has led to the identification of at least five breast cancer subgroups: luminal A, luminal B, normal breast-like, human epidermal growth factor receptor 2, and basal-like. Triple-negative breast cancer is a complex disease diagnosed by immunohistochemistry, and it is characterized by malignant cells not expressing estrogen receptors or progesterone receptors at all, and human epidermal growth factor receptor 2. Along with this knowledge, recent data show that triple-negative breast cancer has specific molecular features that could be possible targets for new biological targeted drugs. The aim of this article is to explore the use of new drugs in this particular setting, which is still associated with poor prognosis and high risk of distant recurrence and death.
Collapse
Affiliation(s)
- Federica Tomao
- Department of Gynecology and Obstetrics, “Sapienza” University of Rome, Policlinico “Umberto I”, Rome, Italy
| | - Anselmo Papa
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Eleonora Zaccarelli
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Luigi Rossi
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Davide Caruso
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Marina Minozzi
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| | - Patrizia Vici
- Division of Medical Oncology B, Regina Elena National Cancer Institute, Rome, Italy
| | - Luigi Frati
- Department of Molecular Medicine, “Sapienza” University of Rome, Policlinico “Umberto I”, Rome, Italy
| | - Silverio Tomao
- Department of Medico-Surgical Sciences and Biotechnologies, “Sapienza” University of Rome, Oncology Unit, Istituto Chirurgico Ortopedico Traumatologico, Latina, Italy
| |
Collapse
|
75
|
Ganesan P, Moulder S, Lee JJ, Janku F, Valero V, Zinner RG, Naing A, Fu S, Tsimberidou AM, Hong D, Stephen B, Stephens P, Yelensky R, Meric-Bernstam F, Kurzrock R, Wheler JJ. Triple-negative breast cancer patients treated at MD Anderson Cancer Center in phase I trials: improved outcomes with combination chemotherapy and targeted agents. Mol Cancer Ther 2014; 13:3175-84. [PMID: 25253784 PMCID: PMC4258414 DOI: 10.1158/1535-7163.mct-14-0358] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) have poor treatment outcomes. We reviewed the electronic records of consecutive patients with metastatic TNBC treated in phase I clinic at MD Anderson Cancer Center (Houston, TX) between Augu st 2005 and May 2012. One hundred and six patients received at least 1 phase I trial. Twelve of 98 evaluable patients (12%) had either complete response (CR; n = 1), partial response (PR; n = 7), or stable disease ≥ 6 months (SD; n = 4). Patients treated on matched therapy (n = 16) compared with those on nonmatched therapy (n = 90) had improved SD ≥ 6 months/PR/CR (33% vs. 8%; P = 0.018) and longer progression-free survival (PFS; median, 6.4 vs. 1.9 months; P = 0.001). Eleven of 57 evaluable patients (19%) treated with combination chemotherapy and targeted therapy had SD ≥ 6 months/PR/CR versus 1 of 41 evaluable patients (2%) treated on other phase I trials (P = 0.013), and longer PFS (3.0 vs. 1.6 months; P < 0.0001). Patients with molecular alterations in the PI3K/AKT/mTOR pathway treated on matched therapy (n = 16) had improved PFS compared with those with and without molecular alterations treated on nonmatched therapy (n = 27; 6.4 vs. 3.2 months; P = 0.036). On multivariate analysis, improved PFS was associated with treatment with combined chemotherapy and targeted agents (P = 0.0002), ≤ 2 metastatic sites (P = 0.003), therapy with PI3K/AKT/mTOR inhibitors for those with cognate pathway abnormalities (P = 0.018), and treatment with antiangiogenic agents (P = 0.023). In summary, combinations of chemotherapy and angiogenesis and/or PI3K/AKT/mTOR inhibitors demonstrated improved outcomes in patients with metastatic TNBC.
Collapse
Affiliation(s)
- Prasanth Ganesan
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stacy Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ralph G Zinner
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Hong
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, The University of California San Diego, La Jolla, California
| | - Jennifer J Wheler
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
76
|
Masuda M, Yamada T. Signaling pathway profiling by reverse-phase protein array for personalized cancer medicine. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1854:651-7. [PMID: 25448010 DOI: 10.1016/j.bbapap.2014.10.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 10/01/2014] [Accepted: 10/20/2014] [Indexed: 11/28/2022]
Abstract
Deregulation of intracellular signaling through accumulation of genetic alterations is a hallmark of cancer. In the past few decades, concerted and systematic efforts have been made to identify key genetic alterations and to develop therapeutic agents targeting active signaling molecules. However, the efficacy of molecular therapeutics often varies among individuals, and precise mapping of active molecules in individual patients is now considered an essential for therapy optimization. Reverse-phase protein array or microarray (RPPA or RPPM) is an emerging antibody-based highly quantitative proteomic technology, especially suitable for profiling of expression and modification of signaling proteins in low abundance. Because the supply of clinical materials is often limited, RPPA technology is highly advantageous for clinical proteomics in view of its high sensitivity as well as accurate quantification. RPPA has now begun to be incorporated into various clinical trials employing molecular-targeted therapeutics. In this article we review and discuss the application of RPPA technology in the fields of basic, preclinical, and clinical research. The RPPA Global Workshop was recently launched to accelerate the exchange of rapidly expanding knowledge of this fascinating technology among academic laboratories and industries worldwide. This article is part of a Special Issue entitled: Medical Proteomics.
Collapse
Affiliation(s)
- Mari Masuda
- Division of Chemotherapy and Clinical Research, Translational Research Group, National Cancer Center Research Institute, Tokyo, Japan
| | - Tesshi Yamada
- Division of Chemotherapy and Clinical Research, Translational Research Group, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
77
|
Zhang H, Cohen AL, Krishnakumar S, Wapnir IL, Veeriah S, Deng G, Coram MA, Piskun CM, Longacre TA, Herrler M, Frimannsson DO, Telli ML, Dirbas FM, Matin AC, Dairkee SH, Larijani B, Glinsky GV, Bild AH, Jeffrey SS. Patient-derived xenografts of triple-negative breast cancer reproduce molecular features of patient tumors and respond to mTOR inhibition. Breast Cancer Res 2014; 16:R36. [PMID: 24708766 PMCID: PMC4053092 DOI: 10.1186/bcr3640] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2013] [Accepted: 03/25/2014] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) is aggressive and lacks targeted therapies. Phosphatidylinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) pathways are frequently activated in TNBC patient tumors at the genome, gene expression and protein levels, and mTOR inhibitors have been shown to inhibit growth in TNBC cell lines. We describe a panel of patient-derived xenografts representing multiple TNBC subtypes and use them to test preclinical drug efficacy of two mTOR inhibitors, sirolimus (rapamycin) and temsirolimus (CCI-779). METHODS We generated a panel of seven patient-derived orthotopic xenografts from six primary TNBC tumors and one metastasis. Patient tumors and corresponding xenografts were compared by histology, immunohistochemistry, array comparative genomic hybridization (aCGH) and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) sequencing; TNBC subtypes were determined. Using a previously published logistic regression approach, we generated a rapamycin response signature from Connectivity Map gene expression data and used it to predict rapamycin sensitivity in 1,401 human breast cancers of different intrinsic subtypes, prompting in vivo testing of mTOR inhibitors and doxorubicin in our TNBC xenografts. RESULTS Patient-derived xenografts recapitulated histology, biomarker expression and global genomic features of patient tumors. Two primary tumors had PIK3CA coding mutations, and five of six primary tumors showed flanking intron single nucleotide polymorphisms (SNPs) with conservation of sequence variations between primary tumors and xenografts, even on subsequent xenograft passages. Gene expression profiling showed that our models represent at least four of six TNBC subtypes. The rapamycin response signature predicted sensitivity for 94% of basal-like breast cancers in a large dataset. Drug testing of mTOR inhibitors in our xenografts showed 77 to 99% growth inhibition, significantly more than doxorubicin; protein phosphorylation studies indicated constitutive activation of the mTOR pathway that decreased with treatment. However, no tumor was completely eradicated. CONCLUSIONS A panel of patient-derived xenograft models covering a spectrum of TNBC subtypes was generated that histologically and genomically matched original patient tumors. Consistent with in silico predictions, mTOR inhibitor testing in our TNBC xenografts showed significant tumor growth inhibition in all, suggesting that mTOR inhibitors can be effective in TNBC, but will require use with additional therapies, warranting investigation of optimal drug combinations.
Collapse
|
78
|
Blanco E, Sangai T, Wu S, Hsiao A, Ruiz-Esparza GU, Gonzalez-Delgado CA, Cara FE, Granados-Principal S, Evans KW, Akcakanat A, Wang Y, Do KA, Meric-Bernstam F, Ferrari M. Colocalized delivery of rapamycin and paclitaxel to tumors enhances synergistic targeting of the PI3K/Akt/mTOR pathway. Mol Ther 2014; 22:1310-1319. [PMID: 24569835 DOI: 10.1038/mt.2014.27] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Accepted: 02/13/2014] [Indexed: 12/19/2022] Open
Abstract
Ongoing clinical trials target the aberrant PI3K/Akt/mammalian target of rapamycin (mTOR) pathway in breast cancer through administration of rapamycin, an allosteric mTOR inhibitor, in combination with paclitaxel. However, synergy may not be fully exploited clinically because of distinct pharmacokinetic parameters of drugs. This study explores the synergistic potential of site-specific, colocalized delivery of rapamycin and paclitaxel through nanoparticle incorporation. Nanoparticle drug loading was accurately controlled, and synergistic drug ratios established in vitro. Precise drug ratios were maintained in tumors 48 hours after nanoparticle administration to mice, at levels twofold greater than liver and spleen, yielding superior antitumor activity compared to controls. Simultaneous and preferential in vivo delivery of rapamycin and paclitaxel to tumors yielded mechanistic insights into synergy involving suppression of feedback loop Akt phosphorylation and its downstream targets. Findings demonstrate that a same time, same place, and specific amount approach to combination chemotherapy by means of nanoparticle delivery has the potential to successfully translate in vitro synergistic findings in vivo. Predictive in vitro models can be used to determine optimum drug ratios for antitumor efficacy, while nanoparticle delivery of combination chemotherapies in preclinical animal models may lead to enhanced understanding of mechanisms of synergy, ultimately opening several avenues for personalized therapy.
Collapse
Affiliation(s)
- Elvin Blanco
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA
| | - Takafumi Sangai
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Suhong Wu
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA
| | - Angela Hsiao
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA
| | - Guillermo U Ruiz-Esparza
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA; Escuela de Biotecnología y Alimentos y Escuela de Medicina, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Carlos A Gonzalez-Delgado
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA; Escuela de Biotecnología y Alimentos y Escuela de Medicina, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey, Mexico
| | - Francisca E Cara
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Kurt W Evans
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA; Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Argun Akcakanat
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ying Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kim-Anh Do
- Department of Biostatistics, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Funda Meric-Bernstam
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, Texas, USA; Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Mauro Ferrari
- Department of Nanomedicine, The Houston Methodist Research Institute, Houston, Texas, USA.
| |
Collapse
|
79
|
Vicier C, Dieci MV, Arnedos M, Delaloge S, Viens P, Andre F. Clinical development of mTOR inhibitors in breast cancer. Breast Cancer Res 2014; 16:203. [PMID: 25189767 PMCID: PMC3978635 DOI: 10.1186/bcr3618] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) pathway is a central pathway that regulates mRNA translation, protein synthesis, glucose metabolism, lipid synthesis and autophagy, and is involved in malignant transformation. Several randomized trials have shown that the use of mTOR inhibitors could improve patient outcome with hormone receptor-positive or human epidermal growth factor receptor-2-positive breast cancer. This review analyzes new perspectives from these trials. Preclinical studies have suggested that the mTOR pathway may play a role in the resistance to hormone therapy, trastuzumab and chemotherapy for breast cancer. This concept has been tested in clinical trials for neoadjuvant treatment and for metastatic breast cancer patients. Also, much effort has gone into the identification of biomarkers that will allow for more precise stratification of patients. Findings from these studies will provide indispensable tools for the design of future clinical trials and identify new perspectives and challenges for researchers and clinicians.
Collapse
|
80
|
Abstract
Triple negative breast cancer (TNBC) is the most lethal form of breast cancer. Treatment options for advanced disease are limited, with a median survival from the time of developing metastases rarely exceeding 1 year. TNBC is heterogeneous, and harbours several molecular alterations. Unfortunately, up to now, clinical trials combining targeted agents and chemotherapy have failed to show substantial survival improvement; therefore, chemotherapy remains the backbone of treatment. No major advances have been made in the field of cytotoxic treatments, and hopefully ongoing trials will contribute to a more precise definition of the role of platinum salts in sporadic and BRCA-mutated TNBC. Moreover, recent gene expression data suggest that TNBC can be further segmented into smaller subgroups, characterized by different activated pathways, which may therefore warrant different targeted treatments. The lack of efficacy that has been observed for the majority of targeted agents in TNBC so far may derive from the inclusion of unselected TNBC patient populations, not enriched for patients presenting an alteration in the target. Therefore, one of the major challenges in the future is to integrate biological data into clinical trials to obtain the highest efficacy from promising targeted treatments such as anti-angiogenetic agents, poly (ADP-ribose) polymerase-1 (PARP), epidermal growth factor receptor, fibroblast growth factor receptor, androgen receptor and phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) inhibitors.
Collapse
|
81
|
Neoadjuvant therapy in operable breast cancer: application to triple negative breast cancer. JOURNAL OF ONCOLOGY 2013; 2013:219869. [PMID: 23983689 PMCID: PMC3747378 DOI: 10.1155/2013/219869] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 07/06/2013] [Indexed: 12/31/2022]
Abstract
Systemic treatment for triple negative breast cancer (TNBC: negative for the expression of estrogen receptor and progesterone receptor and HER2 amplification) has been limited to chemotherapy options. Neoadjuvant chemotherapy induces tumor shrinkage and improves the surgical outcomes of patients with locally advanced disease and also identifies those at high risk of disease relapse despite today's standard of care. By using pathologic complete response as a surrogate endpoint, novel treatment strategies can be efficiently assessed. Tissue analysis in the neoadjuvant setting is also an important research tool for the identification of chemotherapy resistance mechanisms and new therapeutic targets. In this paper, we review data on completed and ongoing neoadjuvant clinical trials in patients with TNBC and discuss treatment controversies that face clinicians and researchers when neoadjuvant chemotherapy is employed.
Collapse
|
82
|
Saksena R, Wong ST. Clinical evidence of the efficacy of everolimus and its potential in the treatment of breast cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2013; 5:27-35. [PMID: 24648755 PMCID: PMC3929343 DOI: 10.2147/bctt.s30131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The PI3K/Akt/mTOR (phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin) pathway regulates several key cellular functions and its dysregulation creates an environment that promotes tumorigenesis as well as resistance to therapy. The mTOR inhibitor everolimus has emerged as a promising agent in the treatment of breast cancer and was recently approved in combination with exemestane for advanced hormone receptor-positive disease after progression on a nonsteroidal aromatase inhibitor. Everolimus may also be effective in combination with cytotoxic and human epidermal growth factor receptor-2-directed therapies for the treatment of other subtypes of breast cancer. This paper highlights preclinical and clinical data that have emerged on the role of mTOR inhibition in breast cancer. Although generally well tolerated, everolimus carries a unique side effect profile of which both patients and providers should be made aware. Recommendations related to the administration of everolimus in the clinical setting are also discussed.
Collapse
Affiliation(s)
- Rujuta Saksena
- The Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Serena T Wong
- The Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
83
|
Amos KD, Adamo B, Anders CK. Triple-negative breast cancer: an update on neoadjuvant clinical trials. Int J Breast Cancer 2012; 2012:385978. [PMID: 22461984 PMCID: PMC3270519 DOI: 10.1155/2012/385978] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 10/13/2011] [Accepted: 10/13/2011] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with a poor prognosis despite the high rates of response to chemotherapy. This scenario highlights the need to develop novel therapies and/or treatment strategies to reduce the mortality associated with TNBC. The neoadjuvant setting provides a model for rapid assessment of treatment efficacy with smaller patient accruals and over shorter periods of time compared to the traditional adjuvant setting. In addition, a clear surrogate endpoint of improved survival, known as pathologic complete response, already exists in this setting. Here, we review current data from completed and ongoing neoadjuvant clinical trials for TNBC.
Collapse
Affiliation(s)
- Keith D. Amos
- Division of Surgical Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Barbara Adamo
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Human Pathology, Integrated Therapies in Oncology Unit, University of Messina, 98125 Messina, Italy
| | - Carey K. Anders
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Hematology/Oncology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|