51
|
Kimura M, Hanamura T, Tsuboi K, Kaneko Y, Yamaguchi Y, Niwa T, Narui K, Endo I, Hayashi SI. Acquired resistance to everolimus in aromatase inhibitor-resistant breast cancer. Oncotarget 2018; 9:21468-21477. [PMID: 29765553 PMCID: PMC5940386 DOI: 10.18632/oncotarget.25133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/23/2018] [Indexed: 12/03/2022] Open
Abstract
We previously reported the establishment of several types of long-term estrogen-depleted-resistant (EDR) cell lines from MCF-7 breast cancer cells. Type 1 EDR cells exhibited the best-studied mechanism of aromatase inhibitor (AI) resistance, in which estrogen receptor (ER) expression remained positive and PI3K signaling was upregulated. Type 2 EDR cells showed reduced ER activity and upregulated JNK-related signaling. The mTOR inhibitor everolimus reduced growth in cells similar to Type 1 EDR cells. The present study generated everolimus-resistant (EvR) cells from Types 1 and 2 EDR cells following long-term exposure to everolimus in vitro. These EvR cells modeled resistance to AI and everolimus combination therapies following first-line AI treatment failure. In Type 1 EvR cells, everolimus resistance was dependent on MAPK signaling; single agents were not effective, but hormonal therapy combined with a kinase inhibitor effectively reduced cell growth. In Type 2 EvR cells, ER expression remained negative and a JNK inhibitor was ineffective, but a Src inhibitor reduced cell growth. The mechanism of acquired everolimus resistance appears to vary depending on the mechanism of AI resistance. Strategies targeting resistant tumors should be tailored based on the resistance mechanisms, as these mechanisms impact therapeutic efficacy.
Collapse
Affiliation(s)
- Mariko Kimura
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Gastroenterological Surgery and Clinical Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.,Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | - Toru Hanamura
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan.,Division of Breast and Endocrine Surgery, Department of Surgery, Shinshu University School of Medicine, Nagano, Japan
| | - Kouki Tsuboi
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yosuke Kaneko
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuri Yamaguchi
- Research Institute for Clinical Oncology, Saitama Cancer Center, Ina-machi, Japan
| | - Toshifumi Niwa
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazutaka Narui
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, Yokohama, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery and Clinical Oncology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Shin-Ichi Hayashi
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
52
|
Clement E, Inuzuka H, Nihira NT, Wei W, Toker A. Skp2-dependent reactivation of AKT drives resistance to PI3K inhibitors. Sci Signal 2018. [PMID: 29535262 DOI: 10.1126/scisignal.aao3810] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The PI3K-AKT kinase signaling pathway is frequently deregulated in human cancers, particularly breast cancer, where amplification and somatic mutations of PIK3CA occur with high frequency in patients. Numerous small-molecule inhibitors targeting both PI3K and AKT are under clinical evaluation, but dose-limiting toxicities and the emergence of resistance limit therapeutic efficacy. Various resistance mechanisms to PI3K inhibitors have been identified, including de novo mutations, feedback activation of AKT, or cross-talk pathways. We found a previously unknown resistance mechanism to PI3K pathway inhibition that results in AKT rebound activation. In a subset of triple-negative breast cancer cell lines, treatment with a PI3K inhibitor or depletion of PIK3CA expression ultimately promoted AKT reactivation in a manner dependent on the E3 ubiquitin ligase Skp2, the kinases IGF-1R (insulin-like growth factor 1 receptor) and PDK-1 (phosphoinositide-dependent kinase-1), and the cell growth and metabolism-regulating complex mTORC2 (mechanistic target of rapamycin complex 2), but was independent of PI3K activity or PIP3 production. Resistance to PI3K inhibitors correlated with the increased abundance of Skp2, ubiquitylation of AKT, cell proliferation in culture, and xenograft tumor growth in mice. These findings reveal a ubiquitin signaling feedback mechanism by which PI3K inhibitor resistance may emerge in aggressive breast cancer cells.
Collapse
Affiliation(s)
- Emilie Clement
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Naoe T Nihira
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alex Toker
- Department of Pathology, Medicine and Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA. .,Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
53
|
Criscitiello C, Viale G, Curigliano G, Goldhirsch A. Profile of buparlisib and its potential in the treatment of breast cancer: evidence to date. BREAST CANCER-TARGETS AND THERAPY 2018; 10:23-29. [PMID: 29430197 PMCID: PMC5796458 DOI: 10.2147/bctt.s134641] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Alteration of the phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin signaling pathway is key for the growth and survival of several cancers, including breast cancer. In addition, dysregulation of PI3K signaling may contribute to resistance to several anticancer agents. PI3K inhibitors may, therefore, be effective as antineoplastic therapy. Buparlisib is a potent and highly specific oral inhibitor of the pan-class I PI3K family. Buparlisib specifically inhibits class I PIK3 in the PI3K/AKT kinase signaling pathway in an ATP-competitive manner, thus inhibiting the production of the secondary messenger phosphatidylinositol (3,4,5)-trisphosphate and activation of the PI3K signaling pathway. This may induce inhibition of tumor cell growth and survival in susceptible tumor cell populations. Buparlisib is currently under investigation in patients with a variety of solid tumors, including breast cancer. Buparlisib has been validated as a promising anticancer agent, and tremendous efforts have been taken to develop it. However, buparlisib monotherapy has resulted in humble benefit so far. Results from studies combining buparlisib with different anticancer agents - namely, endocrine therapy, anti-HER2 therapy, and chemotherapy - have showed variable efficacy with consistent substantial toxicity.
Collapse
|
54
|
Keegan NM, Gleeson JP, Hennessy BT, Morris PG. PI3K inhibition to overcome endocrine resistance in breast cancer. Expert Opin Investig Drugs 2018; 27:1-15. [PMID: 29252036 DOI: 10.1080/13543784.2018.1417384] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Activation of the phosphatidylinositol-3 kinase (PI3K) pathway is a critical step in oncogenesis and plays a role in the development of treatment resistance for both estrogen receptor (ER) positive and human epidermal growth factor receptor 2 (HER2) positive breast cancers. Hence, there have been efforts to therapeutically inhibit this pathway. AREAS COVERED Several inhibitors of PI3K are now progressing through clinical trials with varying degrees of efficacy and toxicity to date. Numerous unresolved questions remain concerning the optimal isoform selectivity of PI3K inhibitors and use of predictive biomarkers. This review examines the most important PI3K inhibitors in ER positive breast cancer to date, with a particular focus on their role in overcoming endocrine therapy resistance and the possible use of PIK3CA mutations as a predictive biomarker. EXPERT OPINION We discuss some of the emerging challenges and questions encountered during the development of PI3K inhibitors from preclinical to phase III studies, including other novel biomarkers and future combinations to overcome endocrine resistance.
Collapse
Affiliation(s)
- Niamh M Keegan
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Jack P Gleeson
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland
| | - Bryan T Hennessy
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| | - Patrick G Morris
- a Department of Medical Oncology , Cancer Clinical Trials and Research Unit, Beaumont Hospital , Dublin , Ireland.,b Department of Molecular Medicine , Royal College of Surgeons in Ireland, Beaumont Hospital , Dublin , Ireland
| |
Collapse
|
55
|
Di Leo A, Johnston S, Lee KS, Ciruelos E, Lønning PE, Janni W, O'Regan R, Mouret-Reynier MA, Kalev D, Egle D, Csőszi T, Bordonaro R, Decker T, Tjan-Heijnen VCG, Blau S, Schirone A, Weber D, El-Hashimy M, Dharan B, Sellami D, Bachelot T. Buparlisib plus fulvestrant in postmenopausal women with hormone-receptor-positive, HER2-negative, advanced breast cancer progressing on or after mTOR inhibition (BELLE-3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 19:87-100. [PMID: 29223745 DOI: 10.1016/s1470-2045(17)30688-5] [Citation(s) in RCA: 295] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/14/2017] [Accepted: 08/16/2017] [Indexed: 01/06/2023]
Abstract
BACKGROUND Activation of the PI3K/AKT/mTOR pathway occurs frequently in breast cancer that is resistant to endocrine therapy. Approved mTOR inhibitors effectively inhibit cell growth and proliferation but elicit AKT phosphorylation via a feedback activation pathway, potentially leading to resistance to mTOR inhibitors. We evaluated the efficacy and safety of buparlisib plus fulvestrant in patients with advanced breast cancer who were pretreated with endocrine therapy and mTOR inhibitors. METHODS BELLE-3 was a randomised, double-blind, placebo-controlled, multicentre, phase 3 study. Postmenopausal women aged 18 years or older with histologically or cytologically confirmed hormone-receptor-positive, HER2-negative, locally advanced or metastatic breast cancer, who had relapsed on or after endocrine therapy and mTOR inhibitors, were recruited from 200 trial centres in 22 countries. Eligible patients were randomly assigned (2:1) via interactive response technology (block size of six) to receive oral buparlisib (100 mg per day) or matching placebo starting on day 1 of cycle 1, plus intramuscular fulvestrant (500 mg) on days 1 and 15 of cycle 1 and on day 1 of subsequent 28-day cycles. Randomisation was stratified by visceral disease status. The primary endpoint was progression-free survival by local investigator assessment as per the Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1 in the full analysis population (all randomised patients, by intention-to-treat). Safety was analysed in all patients who received at least one dose of treatment and at least one post-baseline safety assessment. This study is registered with ClinicalTrials.gov, number NCT01633060, and is ongoing but no longer enrolling patients. FINDINGS Between Jan 15, 2013, and March 31, 2016, 432 patients were randomly assigned to the buparlisib (n=289) or placebo (n=143) groups. Median progression-free survival was significantly longer in the buparlisib versus placebo group (3·9 months [95% CI 2·8-4·2] vs 1·8 months [1·5-2·8]; hazard ratio [HR] 0·67, 95% CI 0·53-0·84, one-sided p=0·00030). The most frequent grade 3-4 adverse events in the buparlisib versus placebo group were elevated alanine aminotransferase (63 [22%] of 288 patients vs four [3%] of 140), elevated aspartate aminotransferase (51 [18%] vs four [3%]), hyperglycaemia (35 [12%] vs none), hypertension (16 [6%] vs six [4%]), and fatigue (ten [3%] vs two [1%]). Serious adverse events were reported in 64 (22%) of 288 patients in the buparlisib group versus 23 (16%) of 140 in the placebo group; the most frequent serious adverse events (affecting ≥2% of patients) were elevated aspartate aminotransferase (six [2%] vs none), dyspnoea (six [2%] vs one [1%]), and pleural effusion (six [2%] vs none). On-treatment deaths occurred in ten (3%) of 288 patients in the buparlisib group and in six (4%) of 140 in the placebo group; most deaths were due to metastatic breast cancer, and two were considered treatment-related (cardiac failure [n=1] in the buparlisib group and unknown reason [n=1] in the placebo group). INTERPRETATION The safety profile of buparlisib plus fulvestrant does not support its further development in this setting. Nonetheless, the efficacy of buparlisib supports the rationale for the use of PI3K inhibitors plus endocrine therapy in patients with PIK3CA mutations. FUNDING Novartis Pharmaceuticals Corporation.
Collapse
Affiliation(s)
- Angelo Di Leo
- Nuovo Ospedale di Prato Santo Stefano, Prato, Italy.
| | | | | | - Eva Ciruelos
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Per E Lønning
- Section of Oncology, Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | | | - Ruth O'Regan
- University of Wisconsin-Madison, Madison, WI, USA
| | | | | | - Daniel Egle
- Universität Frauenklinik Innsbruck, Innsbruck, Austria
| | - Tibor Csőszi
- JNSZ Megyei Hetényi Géza Kórház-Rendelőintézet, Szolnok, Hungary
| | | | | | | | - Sibel Blau
- Rainier Hematology-Oncology/Northwest Medical Specialties, Tacoma, WA, USA
| | - Alessio Schirone
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), Meldola, Italy
| | | | | | - Bharani Dharan
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Dalila Sellami
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | |
Collapse
|
56
|
Vidula N, Rugo HS. Emerging data on improving response to hormone therapy: the role of novel targeted agents. Expert Rev Anticancer Ther 2017; 18:3-18. [PMID: 29192520 DOI: 10.1080/14737140.2018.1412259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Hormone receptor positive (HR+) breast cancer represents the most common subtype of breast cancer. Metastatic HR+ breast cancer may develop resistance to standard hormone therapies, arising from genomic alterations in the estrogen receptor and/or upregulation of other signal transduction pathways. Areas covered: In this review, we discuss hormone resistance and strategies to overcome it, from the pre-clinical and clinical perspectives. This review includes a discussion of inhibition of the PI3K/AKT/mTOR, CDK 4/6, histone deacetylation, fibroblast growth factor receptor, and immune pathways, based on review of relevant literature. Expert commentary: Several emerging novel therapies to improve the response to hormone therapy are approved or are in development. The most promising agents at present are inhibitors of CDK 4/6 and mTOR, which have already been incorporated into treatment in the advanced stage setting and are under study for early stage disease.
Collapse
Affiliation(s)
- Neelima Vidula
- a Division of Hematology and Oncology , Massachusetts General Hospital , Boston , MA , USA
| | - Hope S Rugo
- b Division of Hematology and Oncology , University of California San Francisco, Helen Diller Family Comprehensive Cancer Center , San Francisco , CA , USA
| |
Collapse
|
57
|
Robert M, Frenel JS, Bourbouloux E, Berton Rigaud D, Patsouris A, Augereau P, Gourmelon C, Campone M. Efficacy of buparlisib in treating breast cancer. Expert Opin Pharmacother 2017; 18:2007-2016. [PMID: 29169282 DOI: 10.1080/14656566.2017.1410139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Breast cancer is the most frequent cancer in women. Despite a decline in breast cancer mortality, prognosis of advanced breast cancer remains poor. In a desperate need to improve breast cancer outcomes, newer agents that target molecular pathways are being tested. Deregulation of the phosphoinositide 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) is frequently found in breast cancer. This can lead to resistance of endocrine therapy and anti-HER2 therapies. Targeting this pathway may restore sensitivity to these compounds. Buparlisib (BKM-120) is an orally active pan-PI3K inhibitor evaluated in different tumor types. Areas covered: Buparlisib is one of the most investigated PI3K inhibitors. Preclinical and clinical studies of buparlisib in breast cancer are analyzed and discussed. This article reviews the status of buparlisib, completed and ongoing trials, and its safety. Expert opinion: PI3K inhibitors show promising results in breast cancer. However, we raise a number of issues including the identification of biomarkers to predict treatment response and strategies to counteract resistance. Moreover, its toxicity profile could limit its extensive use.
Collapse
Affiliation(s)
- Marie Robert
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Jean-Sébastien Frenel
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Emmanuelle Bourbouloux
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Dominique Berton Rigaud
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France
| | - Anne Patsouris
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Paule Augereau
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Carole Gourmelon
- b Department of Medical Oncology , Institut de Cancérologie de l'Ouest, Paul Papin , Angers , France
| | - Mario Campone
- a Department of Medical Oncology , Institut de Cancérologie de l'Ouest, René Gauducheau , St. Herblain , France.,c Nantes and Angers , Centre de Recherche en Cancérologie Nantes-Angers (CRNA) , France
| |
Collapse
|
58
|
May CD, Landers SM, Bolshakov S, Ma X, Ingram DR, Kivlin CM, Watson KL, Sannaa GAA, Bhalla AD, Wang WL, Lazar AJ, Torres KE. Co-targeting PI3K, mTOR, and IGF1R with small molecule inhibitors for treating undifferentiated pleomorphic sarcoma. Cancer Biol Ther 2017; 18:816-826. [PMID: 29099264 PMCID: PMC5678691 DOI: 10.1080/15384047.2017.1373230] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Undifferentiated pleomorphic sarcomas (UPSs) are aggressive mesenchymal malignancies with no definitive cell of origin or specific recurrent genetic hallmarks. These tumors are largely chemoresistant; thus, identification of potential therapeutic targets is necessary to improve patient outcome. Previous studies demonstrated that high expression of activated protein kinase B (AKT) in patients with UPS corresponds to poor disease-specific survival. Here, we demonstrate that inhibiting phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) signaling using a small molecule inhibitor reduced UPS cell proliferation and motility and xenograft growth; however, increased phosphorylation of insulin-like growth factor 1 receptor (IGF1R) indicated the potential for adaptive resistance following treatment through compensatory receptor activation. Co-treatment with a dual PI3K/mTOR inhibitor and an anti-IGF1R kinase inhibitor reduced in vivo tumor growth rates despite a lack of antiproliferative effects in vitro. Moreover, this combination treatment significantly decreased UPS cell migration and invasion, which is linked to changes in p27 subcellular localization. Our results demonstrate that targeted inhibition of multiple components of the IGF1R/PI3K/mTOR pathway was more efficacious than single-agent therapy and suggest that co-targeting this pathway could be a beneficial therapeutic strategy for patients with UPS.
Collapse
Affiliation(s)
- Caitlin D May
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| | - Sharon M Landers
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Svetlana Bolshakov
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - XiaoYan Ma
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Davis R Ingram
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Christine M Kivlin
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| | - Kelsey L Watson
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Ghadah A Al Sannaa
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Angela D Bhalla
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Wei-Lien Wang
- c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Alexander J Lazar
- b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA.,c Department of Pathology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Keila E Torres
- a Department of Surgical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b The University of Texas Health Science Center at Houston , Graduate School of Biomedical Sciences , Houston , TX , USA
| |
Collapse
|
59
|
Abstract
Metastasis is one of the most characteristic yet problematic behaviors of cancer cells. Stage IV breast cancer accounts for a large portion of breast cancer-related morbidity and mortality. Despite early detection and improvement in survival owing to advancements in biomedical research and overall improvement of the health system, 6-10% of patients present with stage IV disease in the developed world, with a higher incidence noted elsewhere. Despite advances in biomedical research into cancer, up to 70-80% of patients with stage IV breast cancer die of cancer in 5 years, a disproportionally higher mortality compared with non-metastatic breast cancer. In this article, we review the incidence, survival, heterogeneity, current practice, and challenges in stage IV breast cancer, and we finish by noting new research initiatives to improve poor survival and suggesting future directions. By doing so, we hope to set the basis of future directions for both treating physicians and translational researchers to relieve the suffering of patients with stage IV breast cancer and improve the survival of patients with this dismal disease.
Collapse
Affiliation(s)
- Bora Lim
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Gabriel N Hortobagyi
- Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
60
|
Abstract
Anti-cancer cancer-targeted therapies are designed to exploit a particular vulnerability in the tumor, which in most cases results from its dependence on an oncogene and/or loss of a tumor suppressor. Mutations in the phosphoinositide 3-kinase (PI3K)/AKT/mTOR pathway are freqcuently found in breast cancers and associated with cellular transformation, tumorigenesis, cancer progression, and drug resistance. Several drugs targeting PI3K/ATK/mTOR are currently in clinical trials, mainly in combination with endocrine therapy and anti-HER2 therapy. These drugs are the focus of this review.
Collapse
|
61
|
Araki K, Miyoshi Y. Mechanism of resistance to endocrine therapy in breast cancer: the important role of PI3K/Akt/mTOR in estrogen receptor-positive, HER2-negative breast cancer. Breast Cancer 2017; 25:392-401. [PMID: 29086897 DOI: 10.1007/s12282-017-0812-x] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022]
Abstract
Endocrine therapy is a crucial treatment for estrogen receptor-positive (ER+) breast cancer, with proven clinical benefits. However, adaptive mechanisms emerge in the tumor, causing resistance to endocrine therapy. A better understanding of resistance mechanisms is needed to overcome this problem and to develop new, precise treatment strategies. Accumulating genetic and cancer biological studies demonstrate the importance of understanding the PI3K/Akt/mTOR and CDK4/6/RB pathways in ER+ HER2- breast cancer. PIK3CA (which encodes phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit α) is frequently mutated in breast cancer, and 30% of advanced ER+ HER2- breast cancers have an activating PIK3CA mutation. AKT1 mutations (E17K) have been found in 1.4-8% of breast cancer patients. ER+ breast cancer patients preferentially demonstrate gain of CCND1 (cyclin D1; 58% in luminal B vs. 29% in luminal A) and CDK4 (25% in luminal B vs. 14% in luminal A) and loss of CDKN2A (p16) and CDKN2C (p18), which are negatively regulated with the cell cycle and are correlated with the CDK4/6/RB pathway. Abnormalities in PI3K/Akt/mTOR and CDK4/6/RB pathways due to genetic alterations result in deregulated kinase activity and malignant transformation. This review focuses on the recent reports of the essential role of PI3K/Akt/mTOR and CDK4/6/RB pathways in ER+ HER2- breast cancer.
Collapse
Affiliation(s)
- Kazuhiro Araki
- Division of Breast and Endocrine, Department of Surgery, Cancer Center, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Yasuo Miyoshi
- Division of Breast and Endocrine, Department of Surgery, Cancer Center, Hyogo College of Medicine, 1-1 Mukogawa, Nishinomiya, Hyogo, 663-8501, Japan
| |
Collapse
|
62
|
The Evolving Role of Companion Diagnostics for Breast Cancer in an Era of Next-Generation Omics. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2185-2198. [DOI: 10.1016/j.ajpath.2017.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 02/06/2023]
|
63
|
Allen CE, Laetsch TW, Mody R, Irwin MS, Lim MS, Adamson PC, Seibel NL, Parsons DW, Cho YJ, Janeway K. Target and Agent Prioritization for the Children's Oncology Group-National Cancer Institute Pediatric MATCH Trial. J Natl Cancer Inst 2017; 109:2972640. [PMID: 28376230 DOI: 10.1093/jnci/djw274] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/13/2016] [Indexed: 12/15/2022] Open
Abstract
Over the past decades, outcomes for children with cancer have improved dramatically through serial clinical trials based in large measure on dose intensification of cytotoxic chemotherapy for children with high-risk malignancies. Progress made through such dose intensification, in general, is no longer yielding further improvements in outcome. With the revolution in sequencing technologies and rapid development of drugs that block specific proteins and pathways, there is now an opportunity to improve outcomes for pediatric cancer patients through mutation-based targeted therapeutic strategies. The Children's Oncology Group (COG), in partnership with the National Cancer Institute (NCI), is planning a trial entitled the COG-NCI Pediatric Molecular Analysis for Therapeutic Choice (Pediatric MATCH) protocol utilizing an umbrella design. This protocol will have centralized infrastructure and will consist of a biomarker profiling protocol and multiple single-arm phase II trials of targeted therapies. Pediatric patients with recurrent or refractory solid tumors, lymphomas, or histiocytoses with measurable disease will be eligible. The Pediatric MATCH Target and Agent Prioritization (TAP) committee includes membership representing COG disease committees, the Food and Drug Administration, and the NCI. The TAP Committee systematically reviewed target and agent pairs for inclusion in the Pediatric MATCH trial. Fifteen drug-target pairs were reviewed by the TAP Committee, with seven recommended for further development as initial arms of the Pediatric MATCH trial. The current evidence for availability, efficacy, and safety of targeted agents in children for each class of mutation considered for inclusion in the Pediatric MATCH trial is discussed in this review.
Collapse
Affiliation(s)
- Carl E Allen
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA
| | - Rajen Mody
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | - Meredith S Irwin
- Department of Pediatrics, Division of Hematology/Oncology, Hospital for Sick Children, Toronto, ON, Canada
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Hospital of the University of Pennsylvania and Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter C Adamson
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nita L Seibel
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD, USA
| | - D Williams Parsons
- Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Y Jae Cho
- Division of Pediatric Neurology, Doernbecher Children's Hospital, Portland, OR, USA.,Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Katherine Janeway
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer Center and Blood Disorder Center, Boston, MA, USA
| | | |
Collapse
|
64
|
Baselga J, Im SA, Iwata H, Cortés J, De Laurentiis M, Jiang Z, Arteaga CL, Jonat W, Clemons M, Ito Y, Awada A, Chia S, Jagiełło-Gruszfeld A, Pistilli B, Tseng LM, Hurvitz S, Masuda N, Takahashi M, Vuylsteke P, Hachemi S, Dharan B, Di Tomaso E, Urban P, Massacesi C, Campone M. Buparlisib plus fulvestrant versus placebo plus fulvestrant in postmenopausal, hormone receptor-positive, HER2-negative, advanced breast cancer (BELLE-2): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18:904-916. [PMID: 28576675 DOI: 10.1016/s1470-2045(17)30376-5] [Citation(s) in RCA: 418] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/20/2017] [Accepted: 04/04/2017] [Indexed: 01/09/2023]
Abstract
BACKGROUND Phosphatidylinositol 3-kinase (PI3K) pathway activation is a hallmark of endocrine therapy-resistant, hormone receptor-positive breast cancer. This phase 3 study assessed the efficacy of the pan-PI3K inhibitor buparlisib plus fulvestrant in patients with advanced breast cancer, including an evaluation of the PI3K pathway activation status as a biomarker for clinical benefit. METHODS The BELLE-2 trial was a randomised, double-blind, placebo-controlled, multicentre study. Postmenopausal women aged 18 years or older with histologically confirmed, hormone receptor-positive and human epidermal growth factor (HER2)-negative inoperable locally advanced or metastatic breast cancer whose disease had progressed on or after aromatase inhibitor treatment and had received up to one previous line of chemotherapy for advanced disease were included. Eligible patients were randomly assigned (1:1) using interactive voice response technology (block size of 6) on day 15 of cycle 1 to receive oral buparlisib (100 mg/day) or matching placebo, starting on day 15 of cycle 1, plus intramuscular fulvestrant (500 mg) on days 1 and 15 of cycle 1, and on day 1 of subsequent 28-day cycles. Patients were assigned randomisation numbers with a validated interactive response technology; these numbers were linked to different treatment groups which in turn were linked to treatment numbers. PI3K status in tumour tissue was determined via central laboratory during a 14-day run-in phase. Randomisation was stratified by PI3K pathway activation status (activated vs non-activated vs and unknown) and visceral disease status (present vs absent). Patients, investigators, local radiologists, study team, and anyone involved in the study were masked to the identity of the treatment until unblinding. The primary endpoints were progression-free survival by local investigator assessment per Response Evaluation Criteria In Solid Tumors (version 1.1) in the total population, in patients with known (activated or non-activated) PI3K pathway status, and in PI3K pathway-activated patients. Efficacy analyses were done in the intention-to-treat population. Safety was analysed in all patients who received at least one dose of study drug and had at least one post-baseline safety assessment according to the treatment they received. This trial is registered with ClinicalTrials.gov, number NCT01610284, and is currently ongoing but not recruiting participants. FINDINGS Between Sept 7, 2012, and Sept 10, 2014, 1147 patients from 267 centres in 29 countries were randomly assigned to receive buparlisib (n=576) or placebo plus fulvestrant (n=571). In the total patient population (n=1147), median progression-free survival was 6·9 months (95% CI 6·8-7·8) in the buparlisib group versus 5·0 months (4·0-5·2) in the placebo group (hazard ratio [HR] 0·78 [95% CI 0·67-0·89]; one-sided p=0·00021). In patients with known PI3K status (n=851), median progression-free survival was 6·8 months (95% CI 5·0-7·0) in the buparlisib group vs 4·5 months (3·3-5·0) in the placebo group (HR 0·80 [95% CI 0·68-0·94]; one-sided p=0·0033). In PI3K pathway-activated patients (n=372), median progression-free survival was 6·8 months (95% CI 4·9-7·1) in the buparlisib group versus 4·0 months (3·1-5·2) in the placebo group (HR 0·76 [0·60-0·97], one-sided p=0·014). The most common grade 3-4 adverse events in the buparlisib group versus the placebo group were increased alanine aminotransferase (146 [25%] of 573 patients vs six [1%] of 570), increased aspartate aminotransferase (103 [18%] vs 16 [3%]), hyperglycaemia (88 [15%] vs one [<1%]), and rash (45 [8%] vs none). Serious adverse events were reported in 134 (23%) of 573 patients in the buparlisib group compared with 90 [16%] of 570 patients in the placebo group; the most common serious adverse events (affecting ≥2% of patients) were increased alanine aminotransferase (17 [3%] of 573 vs one [<1%] of 570) and increased aspartate aminotransferase (14 [2%] vs one [<1%]). No treatment-related deaths occurred. INTERPRETATION The results from this study show that PI3K inhibition combined with endocrine therapy is effective in postmenopausal women with endocrine-resistant, hormone receptor-positive and HER2-negative advanced breast cancer. Use of more selective PI3K inhibitors, such as α-specific PI3K inhibitor, is warranted to further improve safety and benefit in this setting. No further studies are being pursued because of the toxicity associated with this combination. FUNDING Novartis Pharmaceuticals Corporation.
Collapse
Affiliation(s)
- José Baselga
- Department of Medicine and Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Seock-Ah Im
- Department of Internal Medicine, Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Hiroji Iwata
- Department of Breast Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Javier Cortés
- Department of Medical Oncology, Ramón y Cajal University Hospital, Madrid, Spain; Vall d'Hebron Institute of Oncology, Barcelona, Spain; Instituto Oncológico Baselga, Hospital Quirónsalud, Barcelona, Spain
| | - Michele De Laurentiis
- Department of Breast Oncology, Istituto Nazionale Tumouri Fondazione G Pascale, Naples, Italy
| | - Zefei Jiang
- Department of Breast Cancer, Beijing 307 Hospital of PLA, Beijing, China
| | - Carlos L Arteaga
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| | - Walter Jonat
- Department of Gynecology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mark Clemons
- Division of Medical Oncology, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Yoshinori Ito
- Department of Radiation Oncology, Cancer Institute Hospital of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ahmad Awada
- Medicine Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Stephen Chia
- Department of Medicine, BC Cancer Agency, Vancouver, BC, Canada
| | - Agnieszka Jagiełło-Gruszfeld
- Department of Breast Cancer and Reconstruction Surgery, Maria Skłodowska Curie Memorial Cancer Centre and Institute of Oncology, Warsaw, Poland
| | | | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, National Yang Ming University, Taipei, Taiwan
| | - Sara Hurvitz
- Division of Haematology-Oncology, Department of Medicine, University of California, Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | - Norikazu Masuda
- Breast Oncology, Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Masato Takahashi
- Department of Breast Surgery, National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Peter Vuylsteke
- Department of Medical Oncology, Université Catholique de Louvain, CHU UCL, Namur, Belgium
| | | | - Bharani Dharan
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | | | | | - Mario Campone
- Institut de Cancérologie de l'Ouest-René Gauducheau Centre de Recherche en Cancérologie, Nantes, France
| |
Collapse
|
65
|
Huang D, Yang F, Wang Y, Guan X. Mechanisms of resistance to selective estrogen receptor down-regulator in metastatic breast cancer. Biochim Biophys Acta Rev Cancer 2017; 1868:148-156. [PMID: 28344099 DOI: 10.1016/j.bbcan.2017.03.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/18/2017] [Accepted: 03/21/2017] [Indexed: 02/07/2023]
Abstract
Based on the prominent role estrogen receptor (ER) plays in breast cancer, endocrine therapy has been developed to block the ER pathway and has shown great effectiveness. Fulvestrant, the first selective ER down-regulator (SERD), was demonstrated to completely suppress ERα and notably efficient. However, resistance to fulvestrant occurs, either intrinsic or acquired during the treatment. Several potential mechanisms inducing fulvestrant resistance have been proposed, composed of activated ERα-independent compensatory growth factor signaling, stimulated downstream kinases, altered cell cycle mediators, etcetera. Experimentally, combinations of fulvestrant with targeted treatments were reported to eliminate the resistance and improve the effect of fulvestrant. Meanwhile, some clinical trials associated with the targeted combination therapies are in progress. This review focuses on the underlying mechanisms that contribute to fulvestrant resistance in ER-positive breast cancer and provides an overview of combined fulvestrant with targeted agents to shed light on optimal therapies for patients with ER-positive breast cancer.
Collapse
Affiliation(s)
- Doudou Huang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Fang Yang
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China
| | - Yucai Wang
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Xiaoxiang Guan
- Department of Medical Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing 210002, PR China.
| |
Collapse
|
66
|
Bohnacker T, Prota AE, Beaufils F, Burke JE, Melone A, Inglis AJ, Rageot D, Sele AM, Cmiljanovic V, Cmiljanovic N, Bargsten K, Aher A, Akhmanova A, Díaz JF, Fabbro D, Zvelebil M, Williams RL, Steinmetz MO, Wymann MP. Deconvolution of Buparlisib's mechanism of action defines specific PI3K and tubulin inhibitors for therapeutic intervention. Nat Commun 2017; 8:14683. [PMID: 28276440 PMCID: PMC5347140 DOI: 10.1038/ncomms14683] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 01/19/2017] [Indexed: 12/17/2022] Open
Abstract
BKM120 (Buparlisib) is one of the most advanced phosphoinositide 3-kinase (PI3K) inhibitors for the treatment of cancer, but it interferes as an off-target effect with microtubule polymerization. Here, we developed two chemical derivatives that differ from BKM120 by only one atom. We show that these minute changes separate the dual activity of BKM120 into discrete PI3K and tubulin inhibitors. Analysis of the compounds cellular growth arrest phenotypes and microtubule dynamics suggest that the antiproliferative activity of BKM120 is mainly due to microtubule-dependent cytotoxicity rather than through inhibition of PI3K. Crystal structures of BKM120 and derivatives in complex with tubulin and PI3K provide insights into the selective mode of action of this class of drugs. Our results raise concerns over BKM120's generally accepted mode of action, and provide a unique mechanistic basis for next-generation PI3K inhibitors with improved safety profiles and flexibility for use in combination therapies. Buparlisib/BKM120 is in phase 3 clinical trials as a phosphoinositide 3-kinase (PI3K) inhibitor. Here, Bohnacker et al. combine chemical biology and structural biology approaches to segregate BKM120's biological actions, and suggest that it causes mitotic arrest predominantly by binding microtubules and disrupting their dynamics.
Collapse
Affiliation(s)
- Thomas Bohnacker
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Andrea E Prota
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen, Switzerland
| | - Florent Beaufils
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia BC V8W 2Y2, Canada
| | - Anna Melone
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | | | - Denise Rageot
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Alexander M Sele
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | | | | | - Katja Bargsten
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen, Switzerland
| | - Amol Aher
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Anna Akhmanova
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - J Fernando Díaz
- CIB Centro de Investigaciones Biológicas, 28040 Madrid, Spain
| | | | | | | | - Michel O Steinmetz
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institut, 5232 Villigen, Switzerland
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| |
Collapse
|
67
|
Oza A, Ma CX. New Insights in Estrogen Receptor (ER) Biology and Implications for Treatment. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0231-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
68
|
Rugo HS, Vidula N, Ma C. Improving Response to Hormone Therapy in Breast Cancer: New Targets, New Therapeutic Options. Am Soc Clin Oncol Educ Book 2017; 35:e40-54. [PMID: 27249746 DOI: 10.1200/edbk_159198] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The majority of breast cancer expresses the estrogen and or progesterone receptors (ER and PR). In tumors without concomitant HER2 amplification, hormone therapy is a major treatment option for all disease stages. Resistance to hormonal therapy is associated with disease recurrence and progression. Recent studies have identified a number of resistance mechanisms leading to estrogen-independent growth of hormone receptor-positive (HR+) breast cancer as a result of genetic and epigenetic alterations, which could be exploited as novel therapeutic targets. These include acquired mutations in ER-alpha (ESR1) in response to endocrine deprivation; constitutive activation of cyclin-dependent kinases (CDK) 4 and 6; cross talk between ER and growth factor receptor signaling such as HER family members, fibroblast growth factor receptor (FGFR) pathways, intracellular growth, and survival signals PI3K/Akt/mTOR; and epigenetic modifications by histone deacetylase (HDAC) as well as interactions with tumor microenvironment and host immune response. Inhibitors of these pathways are being developed to improve efficacy of hormonal therapy for treatment of both metastatic and early-stage disease. Two agents are currently approved in the United States for the treatment of metastatic HR+ breast cancer, including the mTOR inhibitor everolimus and the CDK4/6 inhibitor palbociclib. Management of toxicity is a critical aspect of treatment; the primary toxicity of everolimus is stomatitis (treated with topical steroids) and of palbociclib is neutropenia (treated with dose reduction/delay). Many agents are in clinical trials, primarily in combination with hormone therapy; novel combinations are under active investigation.
Collapse
Affiliation(s)
- Hope S Rugo
- From the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; University of San Francisco School of Medicine, San Francisco, CA; Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Neelima Vidula
- From the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; University of San Francisco School of Medicine, San Francisco, CA; Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Cynthia Ma
- From the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; University of San Francisco School of Medicine, San Francisco, CA; Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO
| |
Collapse
|
69
|
Cotargeting of CYP-19 (aromatase) and emerging, pivotal signalling pathways in metastatic breast cancer. Br J Cancer 2016; 116:10-20. [PMID: 27923036 PMCID: PMC5220158 DOI: 10.1038/bjc.2016.405] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/28/2016] [Accepted: 11/08/2016] [Indexed: 01/08/2023] Open
Abstract
Aromatase inhibition is one of the cornerstones of modern endocrine therapy of oestrogen receptor-positive (ER+) metastatic breast cancer (MBC). The nonsteroidal aromatase inhibitors anastrozole and letrozole, as well as the steroidal aromatase inactivator exemestane, are the preferred drugs and established worldwide in all clinical phases of the disease. However, although many patients suffering from MBC experience an initial stabilisation of their metastatic burden, drug resistance and disease progression occur frequently, following in general only a few months on treatment. Extensive translational research during the past two decades has elucidated the major pathways contributing to endocrine resistance and paved the way for clinical studies investigating the efficacy of novel drug combinations involving aromatase inhibitors and emerging drugable targets like mTOR, PI3K and CDK4/6. The present review summarises the basic research that provided the rationale for new drug combinations involving aromatase inhibitors and the main findings of pivotal clinical trials that have already started to change our way to treat hormone-sensitive MBC. The challenging situation of oestrogen receptor-positive and human epidermal growth factor receptor 2-positive (HER2+) MBC is also shortly reviewed to underline the complexity of the clinical scenario in the heterogeneous subgroups of hormone receptor-positive breast cancer patients and the increasing need for personalised medicine. Finally, we summarise some of the promising findings made with the combination of aromatase inhibitors with other potent endocrine treatment options like fulvestrant, a selective oestrogen receptor downregulator.
Collapse
|
70
|
Epigenetic activation of the prostaglandin receptor EP4 promotes resistance to endocrine therapy for breast cancer. Oncogene 2016; 36:2319-2327. [PMID: 27869171 PMCID: PMC5398938 DOI: 10.1038/onc.2016.397] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 09/07/2016] [Accepted: 09/08/2016] [Indexed: 12/19/2022]
Abstract
Approximately 75% of breast cancers express estrogen receptor α (ERα) and depend on estrogen signals for continued growth. Aromatase inhibitors (AIs) prevent estrogen production and inhibit ER signaling, resulting in decreased cancer recurrence and mortality. Advanced tumors treated with AIs almost always develop resistance to these drugs via the upregulation of alternative growth signals. The mechanisms that drive this resistance-especially epigenetic events that alter gene expression-are, however, not well understood. Genome-wide DNA methylation and expression analysis of cell line models of acquired AI resistance indicated that prostaglandin E2 receptor 4 (PTGER4) is upregulated after demethylation in resistant cells. Knockdown and inhibitor studies demonstrate that PTGER4 is essential for estrogen-independent growth. Our exploratory analysis of downstream signaling indicates that PTGER4 likely promotes AI resistance via ligand-independent activation of the ERα-cofactor CARM1. We believe that we have discovered a novel epigenetic mechanism for altering cell signaling and acquiring endocrine therapy resistance. Our findings indicate that PTGER4 is a potential drug target in AI-resistant cancers. In addition, the epigenetic component of PTGER4 regulation suggests that further study of PTGER4 may yield valuable insights into how DNA methylation-targeted diagnoses and treatments can improve AI-resistant breast cancer treatment.
Collapse
|
71
|
Abstract
The phosphoinositide 3-kinase (PI3K) pathway plays an integral role in many cellular processes and is frequently altered in cancer, contributing to tumor growth and survival. Small molecule inhibitors have been developed that target the three major nodes of this pathway: PI3K, AKT, and mammalian target of rapamycin. However, because oncogenic PI3K pathway activation is achieved in diverse, potentially redundant ways, the clinical efficacy of these inhibitors as monotherapies has, so far, been limited, despite demonstrating promising preclinical activity. Moreover, pathway activation is associated with resistance to other therapies; thus, in combination, PI3K pathway inhibitors could restore therapeutic sensitivity to these agents. To maximize therapeutic benefit, drug combinations and schedules must be explored to identify those with the highest efficacy and lowest toxicity overlap. In addition, defining appropriate patient subpopulations, for both monotherapy and drug combinations, will be important. However, identifying predictive biomarkers remains a challenge.
Collapse
|
72
|
Radhi S. Molecular Changes During Breast Cancer and Mechanisms of Endocrine Therapy Resistance. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:539-562. [PMID: 27865467 DOI: 10.1016/bs.pmbts.2016.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Estrogen receptors (ERs) are expressed in 75% of breast cancers. ERs and their estrogen ligands play a key role in the development and progression of breast cancer. ERs have a genomic activity involving direct modulation of expression of genes vital to cell growth and survival by their classic nuclear receptors. The nongenomic activity is mediated by membrane receptor tyrosine kinases that activate signaling pathways resulting in activation of ER pathway modulators. Endocrine therapies inhibit the growth promoting activity of estrogen. ERs-positive breast cancers can exhibit de novo or acquired endocrine resistance. The mechanisms of endocrine therapy resistance are complex include deregulation of ER pathway, growth factor receptor signaling, cell cycle machinery, and tumor microenvironment. In this chapter, we will review the literature on the biology of ERs, the postulated mechanisms of endocrine therapy resistance, and their clinical implications.
Collapse
Affiliation(s)
- S Radhi
- Texas Tech University Health Science Center, Lubbock, TX, United States.
| |
Collapse
|
73
|
Novel Strategies in Hormone Receptor-Positive Advanced Breast Cancer: Overcoming Endocrine Resistance. CURRENT BREAST CANCER REPORTS 2016. [DOI: 10.1007/s12609-016-0228-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
74
|
Martinello R, Genta S, Galizia D, Geuna E, Milani A, Zucchini G, Valabrega G, Montemurro F. New and developing chemical pharmacotherapy for treating hormone receptor-positive/HER2-negative breast cancer. Expert Opin Pharmacother 2016; 17:2179-2189. [PMID: 27646965 DOI: 10.1080/14656566.2016.1236914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Endocrine therapy is the mainstay of treatment for a substantial proportion of hormone receptor positive (HR+) breast cancer (BC). Indeed, patients with metastatic disease not immediately life threatening may experience long disease control across several lines of endocrine therapy. The major limitation of this therapeutic approach is primary or acquired resistance. A better understanding of endocrine resistance has resulted in newer targeted agents to be added to endocrine therapy. Areas covered: This review highlights new findings in the treatment of HR+/HER2- BC, with a particular focus on new drugs from phase 3 development onwards. Expert opinion: Combining endocrine therapy with agents targeting putative mechanisms of endocrine resistance is a newer treatment paradigm in HR+ BC. Adding a biologically targeted agent to endocrine therapy results in improved response rate, and clinical benefit rate, and prolonged progression-free survival. A clear advantage in overall survival has not yet been reported. Combination therapy allows to delay chemotherapy but increases toxicities and costs, which are critical factors in decision making in the clinical practice. Moreover, identification and validation of biomarkers of response are needed. Ongoing and future trials should elucidate the role of these compounds in the treatment of HR+/HER2- BC.
Collapse
Affiliation(s)
- Rossella Martinello
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| | - Sofia Genta
- b Medical Oncology 1 , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy.,c Department of Oncology , University of Torino , Turin , Italy
| | - Danilo Galizia
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| | - Elena Geuna
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| | - Andrea Milani
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| | - Giorgia Zucchini
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| | - Giorgio Valabrega
- b Medical Oncology 1 , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy.,c Department of Oncology , University of Torino , Turin , Italy
| | - Filippo Montemurro
- a Investigative Clinical Oncology (INCO) , Candiolo Cancer Institute-FPO- IRCCS , Turin , Italy
| |
Collapse
|
75
|
Activation of PI3K/Akt/mTOR signaling in the tumor stroma drives endocrine therapy-dependent breast tumor regression. Oncotarget 2016; 6:22081-97. [PMID: 26098779 PMCID: PMC4673148 DOI: 10.18632/oncotarget.4203] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/27/2015] [Indexed: 12/21/2022] Open
Abstract
Improved efficacy of neoadjuvant endocrine-targeting therapies in luminal breast carcinomas could be achieved with optimal use of pathway targeting agents. In a mouse model of ductal breast carcinoma we identify a tumor regressive stromal reaction that is induced by neoadjuvant endocrine therapy. This reparative reaction is characterized by tumor neovascularization accompanied by infiltration of immune cells and carcinoma-associated fibroblasts that stain for phosphorylated ribosomal protein S6 (pS6), downstream the PI3K/Akt/mTOR pathway. While tumor variants with higher PI3K/Akt/mTOR activity respond well to a combination of endocrine and PI3K/Akt/mTOR inhibitors, tumor variants with lower PI3K/Akt/mTOR activity respond more poorly to the combination therapy than to the endocrine therapy alone, associated with inhibition of stromal pS6 and the reparative reaction. In human breast cancer xenografts we confirm that such differential sensitivity to therapy is primarily determined by the level of PI3K/Akt/mTOR in tumor cells. We further show that the clinical response of breast cancer patients undergoing neoadjuvant endocrine therapy is associated with the reparative stromal reaction. We conclude that tumor level and localization of pS6 are associated with therapeutic response in breast cancer and represent biomarkers to distinguish which tumors will benefit from the incorporation of PI3K/Akt/mTOR inhibitors with neoadjuvant endocrine therapy.
Collapse
|
76
|
Netland IA, Førde HE, Sleire L, Leiss L, Rahman MA, Skeie BS, Miletic H, Enger PØ, Goplen D. Treatment with the PI3K inhibitor buparlisib (NVP-BKM120) suppresses the growth of established patient-derived GBM xenografts and prolongs survival in nude rats. J Neurooncol 2016; 129:57-66. [PMID: 27283525 PMCID: PMC4972854 DOI: 10.1007/s11060-016-2158-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 06/01/2016] [Indexed: 12/03/2022]
Abstract
Glioblastomas (GBMs) are aggressive brain tumours with a dismal prognosis, despite combined surgery, radio- and chemotherapy. Close to 90 % of all GBMs harbour a deregulated PI3K pathway, which is essential in regulating central cellular functions such as proliferation, cell growth, motility and survival. Thus, PI3K represents a potential target for molecular therapy in GBM. We investigated the anti-tumour efficacy of the PI3K inhibitor buparlisib (NVP-BKM120) in GBM cell lines in vitro and in vivo, when treatment was initiated after MRI-confirmed tumour engraftment. We found that buparlisib inhibited glioma cell proliferation in a dose dependent manner, demonstrated by MTS assay, manual cell count and BrdU incorporation. A dose dependent increase in apoptosis was observed through flow cytometric analysis. Furthermore, by immunocytochemistry and western blot, we found a dose dependent inhibition of Akt phosphorylation. Moreover, buparlisib prolonged survival of nude rats harboring human GBM xenografts in three independent studies and reduced the tumours’ volumetric increase, as determined by MRI. In addition, histological analyses of xenograft rat brains showed necrotic areas and change in tumour cell nuclei in buparlisib-treated animals. The rats receiving buparlisib maintained their weight, activity level and food- and water intake. In conclusion, buparlisib effectively inhibits glioma cell proliferation in vitro and growth of human GBM xenografts in nude rats. Moreover, the compound is well tolerated when administered at doses providing anti-tumour efficacy. Thus, buparlisib may have a future role in glioma therapy, and further studies are warranted to validate this compound for human use.
Collapse
Affiliation(s)
- I A Netland
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - H E Førde
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - L Sleire
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - L Leiss
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.,Neuro Clinic, Haukeland University Hospital, Bergen, Norway
| | - M A Rahman
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - B S Skeie
- Department of Clinical Medicine, K1, University of Bergen, Bergen, Norway
| | - H Miletic
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway.,Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - P Ø Enger
- Oncomatrix Research Lab, Department of Biomedicine, University of Bergen, Bergen, Norway.,Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway.,Department of Neurosurgery, Haukeland University Hospital, Bergen, Norway
| | - D Goplen
- Department of Biomedicine, Kristian Gerhard Jebsen Brain Tumour Research Center, University of Bergen, Bergen, Norway. .,Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
77
|
Choudhary V, Gupta S. Comprehensive Gene Mutation Profiling of Breast Tumors: Is It Ready for Prime Time Use? CURRENT BREAST CANCER REPORTS 2016. [DOI: 10.1007/s12609-016-0213-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
78
|
Simioni C, Cani A, Martelli AM, Zauli G, Alameen AAM, Ultimo S, Tabellini G, McCubrey JA, Capitani S, Neri LM. The novel dual PI3K/mTOR inhibitor NVP-BGT226 displays cytotoxic activity in both normoxic and hypoxic hepatocarcinoma cells. Oncotarget 2016; 6:17147-60. [PMID: 26003166 PMCID: PMC4627298 DOI: 10.18632/oncotarget.3940] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 05/05/2015] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common lethal human malignancies worldwide and its advanced status is frequently resistant to conventional chemotherapeutic agents and radiation. We evaluated the cytotoxic effect of the orally bioavailable dual PI3K/mTOR inhibitor, NVP-BGT226, on a panel of HCC cell lines, since hyperactivated PI3K/Akt/mTOR signaling pathway could represent a biomolecular target for Small Inhibitor Molecules in this neoplasia. We analyzed the drug activity in both normoxia and hypoxia conditions, the latter playing often a relevant role in the induction of chemoresistance and angiogenesis.In normoxia NVP-BGT226 caused cell cycle arrest in the G0/G1 phase of the cell cycle, induced apoptosis and autophagy at low concentrations. Interestingly the drug inactivated p-Akt and p-S6 at < 10 nM concentration.In hypoxia NVP-BGT226 maintained its cytotoxic efficacy at the same concentration as documented by MTT assays and Western blot analysis. Moreover, the drug showed in hypoxia inhibitory properties against angiogenesis by lowering the expression of the transcription factor HIF-1α and of VEGF.Our results indicate that NVP-BGT226 has a potent cytotoxic effect on HCC cell lines also in hypoxia condition, thus emerging as a potential candidate for cancer treatment in HCC targeted therapy.
Collapse
Affiliation(s)
- Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Alice Cani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giorgio Zauli
- Institute for Maternal and Child Health, IRCCS "Burlo Garofolo", Trieste, Italy
| | - Ayman A M Alameen
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Department of Chemical Pathology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giovanna Tabellini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy.,LTTA Center, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
79
|
Cristofanilli M, Turner NC, Bondarenko I, Ro J, Im SA, Masuda N, Colleoni M, DeMichele A, Loi S, Verma S, Iwata H, Harbeck N, Zhang K, Theall KP, Jiang Y, Bartlett CH, Koehler M, Slamon D. Fulvestrant plus palbociclib versus fulvestrant plus placebo for treatment of hormone-receptor-positive, HER2-negative metastatic breast cancer that progressed on previous endocrine therapy (PALOMA-3): final analysis of the multicentre, double-blind, phase 3 randomised controlled trial. Lancet Oncol 2016; 17:425-439. [PMID: 26947331 DOI: 10.1016/s1470-2045(15)00613-0] [Citation(s) in RCA: 1269] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 12/11/2015] [Accepted: 12/11/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND In the PALOMA-3 study, the combination of the CDK4 and CDK6 inhibitor palbociclib and fulvestrant was associated with significant improvements in progression-free survival compared with fulvestrant plus placebo in patients with metastatic breast cancer. Identification of patients most suitable for the addition of palbociclib to endocrine therapy after tumour recurrence is crucial for treatment optimisation in metastatic breast cancer. We aimed to confirm our earlier findings with this extended follow-up and show our results for subgroup and biomarker analyses. METHODS In this multicentre, double-blind, randomised phase 3 study, women aged 18 years or older with hormone-receptor-positive, HER2-negative metastatic breast cancer that had progressed on previous endocrine therapy were stratified by sensitivity to previous hormonal therapy, menopausal status, and presence of visceral metastasis at 144 centres in 17 countries. Eligible patients-ie, any menopausal status, Eastern Cooperative Oncology Group performance status 0-1, measurable disease or bone disease only, and disease relapse or progression after previous endocrine therapy for advanced disease during treatment or within 12 months of completion of adjuvant therapy-were randomly assigned (2:1) via a centralised interactive web-based and voice-based randomisation system to receive oral palbociclib (125 mg daily for 3 weeks followed by a week off over 28-day cycles) plus 500 mg fulvestrant (intramuscular injection on days 1 and 15 of cycle 1; then on day 1 of subsequent 28-day cycles) or placebo plus fulvestrant. The primary endpoint was investigator-assessed progression-free survival. Analysis was by intention to treat. We also assessed endocrine therapy resistance by clinical parameters, quantitative hormone-receptor expression, and tumour PIK3CA mutational status in circulating DNA at baseline. This study is registered with ClinicalTrials.gov, NCT01942135. FINDINGS Between Oct 7, 2013, and Aug 26, 2014, 521 patients were randomly assigned, 347 to fulvestrant plus palbociclib and 174 to fulvestrant plus placebo. Study enrolment is closed and overall survival follow-up is in progress. By March 16, 2015, 259 progression-free-survival events had occurred (145 in the fulvestrant plus palbociclib group and 114 in the fulvestrant plus placebo group); median follow-up was 8·9 months (IQR 8·7-9·2). Median progression-free survival was 9·5 months (95% CI 9·2-11·0) in the fulvestrant plus palbociclib group and 4·6 months (3·5-5·6) in the fulvestrant plus placebo group (hazard ratio 0·46, 95% CI 0·36-0·59, p<0·0001). Grade 3 or 4 adverse events occurred in 251 (73%) of 345 patients in the fulvestrant plus palbociclib group and 38 (22%) of 172 patients in the fulvestrant plus placebo group. The most common grade 3 or 4 adverse events were neutropenia (223 [65%] in the fulvestrant plus palbociclib group and one [1%] in the fulvestrant plus placebo group), anaemia (ten [3%] and three [2%]), and leucopenia (95 [28%] and two [1%]). Serious adverse events (all causalities) occurred in 44 patients (13%) of 345 in the fulvestrant plus palbociclib group and 30 (17%) of 172 patients in the fulvestrant plus placebo group. PIK3CA mutation was detected in the plasma DNA of 129 (33%) of 395 patients for whom these data were available. Neither PIK3CA status nor hormone-receptor expression level significantly affected treatment response. INTERPRETATION Fulvestrant plus palbociclib was associated with significant and consistent improvement in progression-free survival compared with fulvestrant plus placebo, irrespective of the degree of endocrine resistance, hormone-receptor expression level, and PIK3CA mutational status. The combination could be considered as a therapeutic option for patients with recurrent hormone-receptor-positive, HER2-negative metastatic breast cancer that has progressed on previous endocrine therapy. FUNDING Pfizer.
Collapse
Affiliation(s)
- Massimo Cristofanilli
- Robert H Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Chicago, IL, USA.
| | | | - Igor Bondarenko
- Dnipropetrovsk Medical Academy, City Multiple-Discipline Clinical Hospital #4, Dnipropetrovsk, Ukraine
| | - Jungsil Ro
- National Cancer Center, Goyang-si, South Korea
| | - Seock-Ah Im
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | | | | | - Angela DeMichele
- Center for Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sherene Loi
- Peter MacCallum Cancer Centre, East Melbourne, VIC, Australia
| | - Sunil Verma
- Sunnybrook Odette Cancer Centre, Toronto, ON, Canada; Tom Baker Cancer Centre, University of Calgary, Calgary, AB, Canada
| | | | - Nadia Harbeck
- Brustzentrum der Universität München (LMU), Munich, Germany
| | | | | | | | | | | | - Dennis Slamon
- University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
80
|
Sini V, Cinieri S, Conte P, De Laurentiis M, Leo AD, Tondini C, Marchetti P. Endocrine therapy in post-menopausal women with metastatic breast cancer: From literature and guidelines to clinical practice. Crit Rev Oncol Hematol 2016; 100:57-68. [PMID: 26944782 DOI: 10.1016/j.critrevonc.2016.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/15/2016] [Indexed: 12/22/2022] Open
Abstract
Current international guidelines recommend endocrine therapy as the initial treatment of choice in hormone receptor positive advanced breast cancer. Endocrine therapy has been a mainstay of hormone responsive breast cancer treatment for more than a century. To date it is based on different approaches,such as blocking the estrogen receptor through selective receptor estrogen modulators, depleting extragonadal peripheral estrogen synthesis by aromatase inhibitors or inducing estrogen receptor degradation using selective down-regulators. Despite estrogen and/or progesterone receptor positive status, up to a quarter of patients could be either primarily resistant to hormone therapies or will develop hormone resistance during the course of their disease. Different mechanisms, either intrinsic or acquired, could be implicated in endocrine resistance. In the present work available endocrine therapies and their appropriate sequences have been reviewed, and the most promising strategies to overcome endocrine resistance have been highlighted.
Collapse
Affiliation(s)
- Valentina Sini
- Surgical and Medical Department of Clinical Sciences, Biomedical Technologies and Translational Medicine, "Sapienza" University of Rome, Italy; Oncology Department, Santo Spirito Hospital, Rome, Italy
| | - Saverio Cinieri
- Medical Oncology Department & Breast Unit-Hospital of Brindisi and Medical Oncology Department-European Institute of Oncology, Milan, Italy
| | - Pierfranco Conte
- Medical Oncology 2, Venetian Oncological Institute, Padova, Italy; Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Michelino De Laurentiis
- Department of Breast Oncology, National Cancer Institute "Fondazione Pascale", Naples, Italy
| | - Angelo Di Leo
- Medical Oncology Department, "Sandro Pitigliani" Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Carlo Tondini
- USC Oncologia Medica, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Medical Oncology Division, Sant'Andrea Hospital, "Sapienza" University of Rome, Rome, Italy; IDI-IRCCS, Rome, Italy.
| |
Collapse
|
81
|
Abstract
Buparlisib (formerly BKM 120), an oral 2,6-dimorpholino pyrimidine derivative is a potent pan-PI3K inhibitor causing inhibition of PI3K downstream signaling including downregulation of p-Akt and p-S6R and apoptosis of cancer cells. Buparlisib is rapidly absorbed, has more than 90% bioavailability, good blood-brain barrier penetration and half-life of 40 h. Phase I trials have shown good disease control rate with tolerable toxicity profile at the recommended doses of 100 mg. The most common adverse events noted with buparlisib are rash, hyperglycemia, derangement of liver functions and psychiatric events. Several clinical trials with buparlisib alone or in combination with chemotherapy and targeted therapies are underway. Buparlisib has not yet been approved for regular use. Further randomized trials are required before buparlisib is approved for treatment of breast cancer.
Collapse
Affiliation(s)
- Bhawna Sirohi
- Department of Medical Oncology, Mazumdar Shaw Cancer Centre, Narayana Health, Bangalore, India
| | | | | |
Collapse
|
82
|
Hudson K, Hancox UJ, Trigwell C, McEwen R, Polanska UM, Nikolaou M, Morentin Gutierrez P, Avivar-Valderas A, Delpuech O, Dudley P, Hanson L, Ellston R, Jones A, Cumberbatch M, Cosulich SC, Ward L, Cruzalegui F, Green S. Intermittent High-Dose Scheduling of AZD8835, a Novel Selective Inhibitor of PI3Kα and PI3Kδ, Demonstrates Treatment Strategies for PIK3CA-Dependent Breast Cancers. Mol Cancer Ther 2016; 15:877-89. [PMID: 26839307 DOI: 10.1158/1535-7163.mct-15-0687] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/25/2016] [Indexed: 11/16/2022]
Abstract
The PIK3CA gene, encoding the p110α catalytic unit of PI3Kα, is one of the most frequently mutated oncogenes in human cancer. Hence, PI3Kα is a target subject to intensive efforts in identifying inhibitors and evaluating their therapeutic potential. Here, we report studies with a novel PI3K inhibitor, AZD8835, currently in phase I clinical evaluation. AZD8835 is a potent inhibitor of PI3Kα and PI3Kδ with selectivity versus PI3Kβ, PI3Kγ, and other kinases that preferentially inhibited growth in cells with mutant PIK3CA status, such as in estrogen receptor-positive (ER(+)) breast cancer cell lines BT474, MCF7, and T47D (sub-μmol/L GI50s). Consistent with this, AZD8835 demonstrated antitumor efficacy in corresponding breast cancer xenograft models when dosed continuously. In addition, an alternative approach of intermittent high-dose scheduling (IHDS) was explored given our observations that higher exposures achieved greater pathway inhibition and induced apoptosis. Indeed, using IHDS, monotherapy AZD8835 was able to induce tumor xenograft regression. Furthermore, AZD8835 IHDS in combination with other targeted therapeutic agents further enhanced antitumor activity (up to 92% regression). Combination partners were prioritized on the basis of our mechanistic insights demonstrating signaling pathway cross-talk, with a focus on targeting interdependent ER and/or CDK4/6 pathways or alternatively a node (mTOR) in the PI3K-pathway, approaches with demonstrated clinical benefit in ER(+) breast cancer patients. In summary, AZD8835 IHDS delivers strong antitumor efficacy in a range of combination settings and provides a promising alternative to continuous dosing to optimize the therapeutic index in patients. Such schedules merit clinical evaluation. Mol Cancer Ther; 15(5); 877-89. ©2016 AACR.
Collapse
Affiliation(s)
- Kevin Hudson
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom.
| | - Urs J Hancox
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Cath Trigwell
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Robert McEwen
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Urszula M Polanska
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Myria Nikolaou
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | | | | | - Oona Delpuech
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Phillippa Dudley
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Lyndsey Hanson
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Rebecca Ellston
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Alys Jones
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Marie Cumberbatch
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Sabina C Cosulich
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Lara Ward
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Francisco Cruzalegui
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Stephen Green
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| |
Collapse
|
83
|
Massihnia D, Perez A, Bazan V, Bronte G, Castiglia M, Fanale D, Barraco N, Cangemi A, Di Piazza F, Calò V, Rizzo S, Cicero G, Pantuso G, Russo A. A headlight on liquid biopsies: a challenging tool for breast cancer management. Tumour Biol 2016; 37:4263-73. [DOI: 10.1007/s13277-016-4856-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/13/2016] [Indexed: 12/16/2022] Open
|
84
|
Ma CX, Sanchez C, Gao F, Crowder R, Naughton M, Pluard T, Creekmore A, Guo Z, Hoog J, Lockhart AC, Doyle A, Erlichman C, Ellis MJ. A Phase I Study of the AKT Inhibitor MK-2206 in Combination with Hormonal Therapy in Postmenopausal Women with Estrogen Receptor-Positive Metastatic Breast Cancer. Clin Cancer Res 2016; 22:2650-8. [PMID: 26783290 DOI: 10.1158/1078-0432.ccr-15-2160] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/02/2016] [Indexed: 01/28/2023]
Abstract
PURPOSE PI3K/AKT pathway activation is an important endocrine resistance mechanism in estrogen receptor-positive (ER(+)) breast cancer. After promising preclinical modeling of MK-2206, an allosteric pan-AKT inhibitor, with either estrogen deprivation or fulvestrant, we conducted a phase I trial in patients with metastatic ER(+)HER2(-) breast cancer to determine the recommended phase II treatment dose (RPTD) of MK-2206 when combined with either anastrozole, fulvestrant, or anastrozole/fulvestrant. EXPERIMENTAL DESIGN ER(+) breast cancer cell lines were exposed in vitro to MK-2206 plus estrogen deprivation with or without fulvestrant and monitored for apoptosis. A standard 3+3 design was employed to first determine the maximum tolerated dose (MTD) of MK-2206 plus anastrozole based on cycle 1 toxicity. Each cycle was 28 days. The RPTD was determined on the basis of toxicities observed at MTD level during the first 3 cycles. Subsequent patients received MK-2206, at the RPTD determined above, plus fulvestrant or anastrozole/fulvestrant to define RPTD for these additional regimens. RESULTS MK-2206 induced apoptosis in parental ER(+) but not in long-term estrogen-deprived cell lines, for which fulvestrant was required for apoptosis induction. Thirty-one patients enrolled. The RPTD was defined as MK-2206 150 mg orally weekly with prednisone prophylaxis for each combination. Grade 3 rash was dose limiting. 42% (95% CI, 23%-63%) patients derived clinical benefit without progression within 6 months. Response was not associated with tumor PIK3CA mutation. CONCLUSIONS MK-2206 plus endocrine treatments were tolerable. MK-2206 in combination with anastrozole is being further evaluated in a phase II neoadjuvant trial for newly diagnosed ER(+)HER2(-) breast cancer. Clin Cancer Res; 22(11); 2650-8. ©2016 AACRSee related commentary by Jansen et al., p. 2599.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Cesar Sanchez
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert Crowder
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Naughton
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy Pluard
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Allison Creekmore
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zhanfang Guo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - A Craig Lockhart
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Austin Doyle
- Cancer Therapy Evaluation Program, NCI, Bethesda, MD
| | | | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
85
|
Ma CX, Bose R, Ellis MJ. Prognostic and Predictive Biomarkers of Endocrine Responsiveness for Estrogen Receptor Positive Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 882:125-54. [PMID: 26987533 DOI: 10.1007/978-3-319-22909-6_5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The estrogen-dependent nature of breast cancer is the fundamental basis for endocrine therapy. The presence of estrogen receptor (ER), the therapeutic target of endocrine therapy, is a prerequisite for this therapeutic approach. However, estrogen-independent growth often exists de novo at diagnosis or develops during the course of endocrine therapy. Therefore ER alone is insufficient in predicting endocrine therapy efficacy. Several RNA-based multigene assays are now available in clinical practice to assess distant recurrence risk, with majority of these assays evaluated in patients treated with 5 years of adjuvant endocrine therapy. While MammaPrint and Oncotype Dx are most predictive of recurrence risk within the first 5 years of diagnosis, Prosigna, Breast Cancer Index (BCI), and EndoPredict Clin have also demonstrated utility in predicting late recurrence. In addition, PAM50, or Prosigna, provides further biological insights by classifying breast cancers into intrinsic molecular subtypes. Additional strategies are under investigation in prospective clinical trials to differentiate endocrine sensitive and resistant tumors and include on-treatment Ki-67 and Preoperative Endocrine Prognostic Index (PEPI) score in the setting of neoadjuvant endocrine therapy. These biomarkers have become important tools in clinical practice for the identification of low risk patients for whom chemotherapy could be avoided. However, there is much work ahead toward the development of a molecular classification that informs the biology and novel therapeutic targets in high-risk disease as chemotherapy has only modest benefit in this population. The recognition of somatic mutations and their relationship to endocrine therapy responsiveness opens important opportunities toward this goal.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, 660 South Euclid Avenue, 63110, St Louis, MO, USA
| | - Ron Bose
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, 660 South Euclid Avenue, 63110, St Louis, MO, USA
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, 320A Cullen, MS 600, 77030, Houston, TX, USA.
| |
Collapse
|
86
|
Brufsky AM. Delaying Chemotherapy in the Treatment of Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Advanced Breast Cancer. Clin Med Insights Oncol 2015; 9:137-47. [PMID: 26793013 PMCID: PMC4697769 DOI: 10.4137/cmo.s31586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 10/27/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022] Open
Abstract
Global guidelines for the management of locally advanced or metastatic hormone receptor-positive (HR-positive), human epidermal growth factor 2-negative (HER2-negative) breast cancer recommend endocrine therapy as first-line treatment for all patients, regardless of age or postmenopausal status. However, current practice patterns in the United States and Europe suggest that these modes of therapy are not being used as recommended, and many patients with advanced HR-positive, HER2-negative disease are being treated first-line with chemotherapy or switched to chemotherapy after a single endocrine therapy. Given that chemotherapy is associated with increased toxicity and reduced quality of life (QOL) compared with endocrine therapy, prolonging the duration of response obtained with endocrine therapy may help delay chemotherapy and its attendant toxicities. Several strategies to delay or overcome endocrine resistance and thereby postpone chemotherapy have been explored, including the use of second-line endocrine agents with different mechanisms of action, adding targeted agents that inhibit specific resistance pathways, and adding agents that act in complementary or synergistic ways to inhibit tumor cell proliferation. This review analyzes the different therapy options available to HR-positive, HER2-negative patients with advanced breast cancer that can be used to delay chemotherapy and enhance QOL.
Collapse
Affiliation(s)
- Adam M. Brufsky
- Professor of Medicine, Associate Division Chief of Hematology/Oncology, Medical Director of Women’s Cancer Center at Magee-Womens Hospital, Codirector of Comprehensive Breast Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
87
|
Nguyen VTM, Barozzi I, Faronato M, Lombardo Y, Steel JH, Patel N, Darbre P, Castellano L, Győrffy B, Woodley L, Meira A, Patten DK, Vircillo V, Periyasamy M, Ali S, Frige G, Minucci S, Coombes RC, Magnani L. Differential epigenetic reprogramming in response to specific endocrine therapies promotes cholesterol biosynthesis and cellular invasion. Nat Commun 2015; 6:10044. [PMID: 26610607 PMCID: PMC4674692 DOI: 10.1038/ncomms10044] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 10/28/2015] [Indexed: 12/24/2022] Open
Abstract
Endocrine therapies target the activation of the oestrogen receptor alpha (ERα) via distinct mechanisms, but it is not clear whether breast cancer cells can adapt to treatment using drug-specific mechanisms. Here we demonstrate that resistance emerges via drug-specific epigenetic reprogramming. Resistant cells display a spectrum of phenotypical changes with invasive phenotypes evolving in lines resistant to the aromatase inhibitor (AI). Orthogonal genomics analysis of reprogrammed regulatory regions identifies individual drug-induced epigenetic states involving large topologically associating domains (TADs) and the activation of super-enhancers. AI-resistant cells activate endogenous cholesterol biosynthesis (CB) through stable epigenetic activation in vitro and in vivo. Mechanistically, CB sparks the constitutive activation of oestrogen receptors alpha (ERα) in AI-resistant cells, partly via the biosynthesis of 27-hydroxycholesterol. By targeting CB using statins, ERα binding is reduced and cell invasion is prevented. Epigenomic-led stratification can predict resistance to AI in a subset of ERα-positive patients.
Collapse
Affiliation(s)
- Van T. M. Nguyen
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Iros Barozzi
- IFOM-IEO Campus, European Institute of Oncology, Milan 20139, Italy
| | - Monica Faronato
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Ylenia Lombardo
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Jennifer H. Steel
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Naina Patel
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Philippa Darbre
- School of Biological Science, University of Reading, Reading RG6 6LA, UK
| | - Leandro Castellano
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Balázs Győrffy
- MTA TTK Lendület Cancer Biomarker Research Group, 2nd Department of Pediatrics, Semmelweis University, Budapest H-1117, Hungary
- MTA-SE Pediatrics and Nephrology Research Group, 2nd Department of Pediatrics, Semmelweis University, Budapest H-1117, Hungary
| | | | - Alba Meira
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Darren K. Patten
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Valentina Vircillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende (CS) 87036, Italy
| | | | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Gianmaria Frige
- IFOM-IEO Campus, European Institute of Oncology, Milan 20139, Italy
| | - Saverio Minucci
- IFOM-IEO Campus, European Institute of Oncology, Milan 20139, Italy
| | - R. Charles Coombes
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| | - Luca Magnani
- Department of Surgery and Cancer, Imperial College London, London W12 0NN, UK
| |
Collapse
|
88
|
Ma CX, Luo J, Naughton M, Ademuyiwa F, Suresh R, Griffith M, Griffith OL, Skidmore ZL, Spies NC, Ramu A, Trani L, Pluard T, Nagaraj G, Thomas S, Guo Z, Hoog J, Han J, Mardis E, Lockhart C, Ellis MJ. A Phase I Trial of BKM120 (Buparlisib) in Combination with Fulvestrant in Postmenopausal Women with Estrogen Receptor-Positive Metastatic Breast Cancer. Clin Cancer Res 2015; 22:1583-91. [PMID: 26563128 DOI: 10.1158/1078-0432.ccr-15-1745] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 09/27/2015] [Indexed: 12/25/2022]
Abstract
PURPOSE This trial was conducted to determine the maximum tolerated dose (MTD) and preliminary efficacy of buparlisib, an oral pan-class I PI3K inhibitor, plus fulvestrant in postmenopausal women with metastatic estrogen receptor positive (ER(+)) breast cancer. EXPERIMENTAL DESIGN Phase IA employed a 3+3 design to determine the MTD of buparlisib daily plus fulvestrant. Subsequent cohorts (phase IB and cohort C) evaluated intermittent (5/7-day) and continuous dosing of buparlisib (100 mg daily). No more than 3 prior systemic treatments in the metastatic setting were allowed in these subsequent cohorts. RESULTS Thirty-one patients were enrolled. MTD was defined as buparlisib 100 mg daily plus fulvestrant. Common adverse events (AE) included fatigue (38.7%), transaminases elevation (35.5%), rash (29%), and diarrhea (19.4%). C-peptide was significantly increased during treatment, consistent with on-target effect of buparlisib. Compared with intermittent dosing, daily buparlisib was associated with more frequent early onset AEs and higher buparlisib plasma concentrations. Among the 29 evaluable patients, the clinical benefit rate was 58.6% (95% CI, 40.7%-74.5%). Response was not associated with PIK3CA mutation or treatment cohort; however, loss of PTEN, progesterone receptor (PgR) expression, or mutation in TP53 was most common in resistant cases, and mutations inAKT1 and ESR1 did not exclude treatment response. CONCLUSIONS Buparlisib plus fulvestrant is clinically active with manageable AEs in patients with metastatic ER(+)breast cancer. Weekend breaks in buparlisib dosing reduced toxicity. Patients with PgR negative and TP53 mutation did poorly, suggesting buparlisib plus fulvestrant may not be adequately effective against tumors with these poor prognostic molecular features.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri.
| | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Naughton
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Foluso Ademuyiwa
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Rama Suresh
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Malachi Griffith
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri. The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri. Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Obi L Griffith
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri. The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri. Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Zachary L Skidmore
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Nicholas C Spies
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Avinash Ramu
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Lee Trani
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy Pluard
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Gayathri Nagaraj
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Shana Thomas
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zhanfang Guo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jing Han
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Elaine Mardis
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri. The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri. Department of Genetics, Washington University School of Medicine, St. Louis, Missouri. Division of Genomics and Bioinformatics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Craig Lockhart
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri. Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
89
|
Reinert T, Barrios CH. Optimal management of hormone receptor positive metastatic breast cancer in 2016. Ther Adv Med Oncol 2015; 7:304-20. [PMID: 26557899 PMCID: PMC4622303 DOI: 10.1177/1758834015608993] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hormone receptor positive tumors represent the most common form of breast cancer and account for most of the deaths from the disease. Endocrine therapy represents the main initial therapeutic strategy for these patients and has been associated with significant clinical benefits in a majority of patients. While in early stages endocrine therapy is administered as part of a curative approach once clinical metastases develop, the disease is considered incurable and the main management objectives are tumor control and quality of life. The two major clinical paradigms of always indicating endocrine therapy in the absence of visceral crises and sequencing endocrine treatments have been guiding our therapeutic approach to these patients. However, for many decades, we have delivered endocrine therapy with a 'one size fits all' approach by applying agents that interfere with hormone receptor signaling equally in every clinical patient scenario. We have been unable to incorporate the well-known biologic principle of different degrees of hormone receptor dependency in our therapeutic recommendations. Recent developments in the understanding of molecular interactions of hormone signaling with other important growth factor, metabolic and cell division pathways have opened the possibility of improving results by modulating hormone signaling and interfering with resistance mechanisms yet to be fully understood. Unfortunately, limitations in the design of trials conducted in this area have made it difficult to develop predictive biomarkers and most of the new combinations with targeted agents, even though showing improvements in clinical endpoints, have been directed to an unselected population of patients. In this review we explore some of the current and most relevant literature in the management of hormone receptor positive advance breast cancer.
Collapse
Affiliation(s)
- Tomas Reinert
- Instituto do Câncer, Sistema de Saúde Mãe de Deus, Porto Alegre, RS, Brazil
| | - Carlos H. Barrios
- PUCRS School of Medicine, Department of Medicine, Padre Chagas 66/203, CEP 90 570 080, Porto Alegre, RS, Brazil
| |
Collapse
|
90
|
Estévez LG, García E, Hidalgo M. Inhibiting the PI3K signaling pathway: buparlisib as a new targeted option in breast carcinoma. Clin Transl Oncol 2015; 18:541-9. [PMID: 26510854 DOI: 10.1007/s12094-015-1410-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/14/2015] [Indexed: 01/13/2023]
Abstract
Aberrations in the PI3K signaling pathway are frequently observed in patients with breast cancer. Because of that, PI3K inhibitors are attractive options for the treatment of breast cancer because PI3K is the most proximal component of the pathway other than receptor tyrosine kinases. Buparlisib is a potent and highly specific oral pan-class I PI3K inhibitor, which is currently under investigation in patients with breast cancer. In this article, we describe the PI3K signaling pathway, the prognostic value of PI3K pathway mutations, as well as the mechanism of action of buparlisib. Lastly, we discuss preliminary results of preclinical and clinical studies showing the efficacy and safety profile of this agent in breast cancer patients.
Collapse
Affiliation(s)
- L G Estévez
- Breast Cancer Programme, Centro Integral Oncológico Clara Campal, Oña, 10, 28050, Madrid, Spain.
| | - E García
- Pathology Department, Hospital Universitario Fundación Alcorcón, Budapest, 1, 28922, Alcorcón, Madrid, Spain
| | - M Hidalgo
- Clinical Research Programme, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| |
Collapse
|
91
|
Riseberg D. Treating Elderly Patients With Hormone Receptor-Positive Advanced Breast Cancer. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2015; 9:65-73. [PMID: 26339192 PMCID: PMC4550185 DOI: 10.4137/cmo.s26067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 06/18/2015] [Accepted: 06/20/2015] [Indexed: 02/06/2023]
Abstract
As the overall population ages, the proportion of elderly patients (aged ≥65 years) with breast cancer also increases. Studies have shown that elderly patients with hormone receptor–positive breast cancer can derive as much benefit from treatment as do younger patients, yet they remain underrepresented in clinical trials and are often undertreated in clinical practice. Treatment decisions for older patients should not be based solely on chronologic age; a patient’s physiologic functioning and comorbidities must also be taken into consideration. For recurrent or metastatic disease, systemic treatment with endocrine therapies or chemotherapy may prolong a patient’s life and alleviate troublesome symptoms. Resistance to therapy remains a problem in the advanced breast cancer setting, with most patients eventually becoming resistant to additional treatment. New combination regimens that target multiple pathways, such as everolimus plus exemestane, have shown efficacy in elderly patients previously resistant to endocrine therapies, and future research may need to focus on such combinations in order to improve outcomes in this patient group. A number of investigational agents are in clinical development, although few studies identify their effects in the elderly patient population. Optimizing effective yet tolerable therapeutic regimens for elderly patients could improve their outcomes while ensuring that the goals of improved survival and quality of life are considered.
Collapse
Affiliation(s)
- David Riseberg
- Institute for Cancer Care, Mercy Medical Center, Baltimore, MD, USA
| |
Collapse
|
92
|
Teo ZL, Loi S. Molecular segmentation of luminal breast: Reality in 2015? Breast 2015; 24 Suppl 2:S41-3. [PMID: 26255199 DOI: 10.1016/j.breast.2015.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PURPOSE With the advent of next generation sequencing, we are beginning to get a clearer picture of the landscape of genetic alterations in primary breast cancer. One of the key findings has been the heterogeneity of mutational profiles, with no two primary breast tumors being alike. How these genetic alterations will help us manage patients is currently unclear. Studies are ongoing to determine which genetic alterations will signify "oncogenic addiction" and which ones will determine resistance and relapse to current standard therapies.
Collapse
Affiliation(s)
- Zhi Ling Teo
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - Sherene Loi
- Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia.
| |
Collapse
|
93
|
Samartzis EP, Gutsche K, Dedes KJ, Fink D, Stucki M, Imesch P. Loss of ARID1A expression sensitizes cancer cells to PI3K- and AKT-inhibition. Oncotarget 2015; 5:5295-303. [PMID: 24979463 PMCID: PMC4170604 DOI: 10.18632/oncotarget.2092] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
ARID1A mutations are observed in various tumors, including ovarian clear cell (OCCC) and endometrioid carcinomas, endometrial, and breast carcinomas. They commonly result in loss of ARID1A-protein expression and frequently co-occur with PI3K/AKT-pathway activating mechanisms. The aim of this study was to test the hypothesis as to whether PI3K/AKT-pathway activation is a critical mechanism in ARID1A-mutated tumors and if consequently ARID1A-deficient tumors show increased sensitivity to treatment with PI3K- and AKT-inhibitors. Upon ARID1A knockdown, MCF7 breast cancer cells and primary MRC5 cells exhibited a significantly increased sensitivity towards the AKT-inhibitors MK-2206 and perifosine, as well as the PI3K-inhibitor buparlisib. Knockdown of ARID1A in MCF7 led to an increase of pAKT-Ser473. AKT-inhibition with MK-2206 led to increased apoptosis and to a decrease of pS6K in ARID1A-depleted MCF7 cells but not in the controls. In five OCCC cell lines ARID1A-deficiency correlated with increased pAKT-Ser473 levels and with sensitivity towards treatment with the AKT-inhibitor MK-2206. In conclusion, ARID1A-deficient cancer cells demonstrate an increased sensitivity to treatment with small molecule inhibitors of the PI3K/AKT-pathway. These findings suggest a specific requirement of the PI3K/AKT pathway in ARID1A-deficient tumors and reveal a synthetic lethal interaction between loss of ARID1A expression and inhibition of the PI3K/AKT pathway.
Collapse
Affiliation(s)
| | - Katrin Gutsche
- Department of Gynecology, University Hospital of Zurich, Zürich, Switzerland
| | - Konstantin J Dedes
- Department of Gynecology, University Hospital of Zurich, Zürich, Switzerland
| | - Daniel Fink
- Department of Gynecology, University Hospital of Zurich, Zürich, Switzerland
| | - Manuel Stucki
- Department of Gynecology, University Hospital of Zurich, Zürich, Switzerland
| | - Patrick Imesch
- Department of Gynecology, University Hospital of Zurich, Zürich, Switzerland
| |
Collapse
|
94
|
Wheler JJ, Atkins JT, Janku F, Moulder SL, Yelensky R, Stephens PJ, Kurzrock R. Multiple gene aberrations and breast cancer: lessons from super-responders. BMC Cancer 2015; 15:442. [PMID: 26021831 PMCID: PMC4446801 DOI: 10.1186/s12885-015-1439-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 05/14/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The presence of multiple molecular aberrations in patients with breast cancer may correlate with worse outcomes. CASE PRESENTATIONS We performed in-depth molecular analysis of patients with estrogen receptor-positive, HER2-negative, hormone therapy-refractory breast cancer, who achieved partial or complete responses when treated with anastrozole and everolimus. Tumors were analyzed using a targeted next generation sequencing (NGS) assay in a Clinical Laboratory Improvement Amendments laboratory. Genomic libraries were captured for 3,230 exons in 182 cancer-related genes plus 37 introns from 14 genes often rearranged in cancer and sequenced to high coverage. Patients received anastrozole (1 g PO daily) and everolimus (5 or 10 mg PO daily). Thirty-two patients with breast cancer were treated on study and 5 (16 %) achieved a partial or complete response. Primary breast tissue was available for NGS testing in three of the responders (partial response with progression free survival of 11 and 14 months, respectively; complete response with progression free survival of 9+ months). The following molecular aberrations were observed: PTEN loss by immunohistochemistry, CCDN1 and FGFR1 amplifications, and PRKDC re-arrangement (NGS) (patient #1); PIK3CA and PIK3R1 mutations, and CCDN1, FGFR1, MYC amplifications (patient #2); TP53 mutation, CCNE1, IRS2 and MCL1 amplifications (patient #3). Some (but not all) of these aberrations converge on the PI3K/AKT/mTOR pathway, perhaps accounting for response. CONCLUSIONS Patients with estrogen receptor-positive breast cancer can achieve significant responses on a combination of anastrozole and everolimus, even in the presence of multiple molecular aberrations. Further study of next generation sequencing-profiled tumors for convergence and resistance pathways is warranted.
Collapse
Affiliation(s)
- Jennifer J Wheler
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Box 0455, Houston, TX, 77030, USA.
| | - Johnique T Atkins
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Box 0455, Houston, TX, 77030, USA.
| | - Filip Janku
- Department of Investigational Cancer Therapeutics (Phase I Program), The University of Texas MD Anderson Cancer Center, 1400 Holcombe Boulevard, Box 0455, Houston, TX, 77030, USA.
| | - Stacy L Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | | | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, University of California at San Diego Moores Cancer Center, La Jolla, CA, USA.
| |
Collapse
|
95
|
Abstract
Oestrogen receptor-positive (ER(+)) breast cancer is a major cause of cancer death in women. Although aromatase inhibitors suppress the function of ER and reduce the risk of recurrence, therapeutic resistance is common and essentially inevitable in advanced disease. This Review considers both genomic and cell biological explanations as to why ER(+) breast cancer cells persist, progress and cause an incurable, lethal, systemic disease. The design and outcomes of clinical trials are considered with the perspective that resistance mechanisms are heterogeneous, and therefore biomarker and somatic mutation-based stratification and eligibility will be essential for improvements in patient outcomes.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, Missouri 63110, USA
| | - Tomás Reinert
- Department of Medical Oncology, Instituto Nacional de Câncer (INCA), Praça da Cruz Vermelha, 23, 20230-130, Rio de Janeiro, Brazil
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8 St., 20-954, Lublin, Poland
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston 77030, Texas, USA
| |
Collapse
|
96
|
Jordan VC, Curpan R, Maximov PY. Estrogen receptor mutations found in breast cancer metastases integrated with the molecular pharmacology of selective ER modulators. J Natl Cancer Inst 2015; 107:djv075. [PMID: 25838462 DOI: 10.1093/jnci/djv075] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2014] [Accepted: 02/24/2015] [Indexed: 01/11/2023] Open
Abstract
The consistent reports of mutations at Asp538 and Tyr537 in helix 12 of the ligand-binding domain (LBD) of estrogen receptors (ERs) from antihormone-resistant breast cancer metastases constitute an important advance. The mutant amino acids interact with an anchor amino acid, Asp351, to close the LBD, thereby creating a ligand-free constitutively activated ER. Amino acids Asp 538, Tyr 537, and Asp 351 are known to play a role in either the turnover of ER, the antiestrogenic activity of the ER complex, or the estrogen-like actions of selective ER modulators. A unifying mechanism of action for these amino acids to enhance ER gene activation and growth response is presented. There is a range of mutations described in metastases vs low to zero in primary disease, so the new knowledge is of clinical relevance, thereby confirming an additional mechanism of acquired resistance to antihormone therapy through cell population selection pressure and enrichment during treatment. Circulating tumor cells containing ER mutations can be cultured ex vivo, and tumor tissues can be grown as patient-derived xenografts to add a new dimension for testing drug susceptibility for future drug discovery.
Collapse
Affiliation(s)
- V Craig Jordan
- MD Anderson Cancer Center, Department of Breast Medical Oncology, Houston, TX (VCJ, PYM); Institute of Chemistry, Romanian Academy, Timisoara, Romania (RC).
| | - Ramona Curpan
- MD Anderson Cancer Center, Department of Breast Medical Oncology, Houston, TX (VCJ, PYM); Institute of Chemistry, Romanian Academy, Timisoara, Romania (RC)
| | - Philipp Y Maximov
- MD Anderson Cancer Center, Department of Breast Medical Oncology, Houston, TX (VCJ, PYM); Institute of Chemistry, Romanian Academy, Timisoara, Romania (RC)
| |
Collapse
|
97
|
Xue B, Huang W, Yuan X, Xu B, Lou Y, Zhou Q, Ran F, Ge Z, Li R, Cui J. YSY01A, a Novel Proteasome Inhibitor, Induces Cell Cycle Arrest on G2 Phase in MCF-7 Cells via ERα and PI3K/Akt Pathways. J Cancer 2015; 6:319-26. [PMID: 25767601 PMCID: PMC4349871 DOI: 10.7150/jca.10733] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/15/2014] [Indexed: 11/05/2022] Open
Abstract
Given that the proteasome is essential for multiple cellular processes by degrading diverse regulatory proteins, inhibition of the proteasome has emerged as an attractive target for anti-cancer therapy. YSY01A is a novel small molecule compound targeting the proteasome. The compound was found to suppress viability of MCF-7 cells and cause limited cell membrane damage as determined by sulforhodamine B assay (SRB) and CytoTox 96(®) non-radioactive cytotoxicity assay. High-content screening (HCS) further shows that YSY01A treatment induces cell cycle arrest on G2 phase within 24 hrs. Label-free quantitative proteomics (LFQP), which allows extensive comparison of cellular responses following YSY01A treatment, suggests that various regulatory proteins including cell cycle associated proteins and PI3K/Akt pathway may be affected. Furthermore, YSY01A increases p-CDC-2, p-FOXO3a, p53, p21(Cip1) and p27(Kip1) but decreases p-Akt, p-ERα as confirmed by Western blotting. Therefore, YSY01A represents a potential therapeutic for breast cancer MCF-7 by inducing G2 phase arrest via ERα and PI3K/Akt pathways.
Collapse
Affiliation(s)
- Bingjie Xue
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Wei Huang
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Xia Yuan
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Bo Xu
- 2. Instrumental Analysis Center of State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Yaxin Lou
- 3. Lab of Proteomics Medical and Healthy Analytical Center, Peking University, Beijing, China
| | - Quan Zhou
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Fuxiang Ran
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| | - Zemei Ge
- 4. Peking University School of Pharmaceutical Sciences Department of Medicinal Chemistry, Beijing, China
| | - Runtao Li
- 4. Peking University School of Pharmaceutical Sciences Department of Medicinal Chemistry, Beijing, China
| | - Jingrong Cui
- 1. State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100083, Beijing, China
| |
Collapse
|
98
|
Geuna E, Milani A, Martinello R, Aversa C, Valabrega G, Scaltriti M, Montemurro F. Buparlisib , an oral pan-PI3K inhibitor for the treatment of breast cancer. Expert Opin Investig Drugs 2015; 24:421-31. [PMID: 25645727 DOI: 10.1517/13543784.2015.1008132] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Deregulation of the phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) intracellular signaling pathway is common in breast cancer (BC) and has been found to be potentially implicated in resistance to endocrine and anti-HER2 therapies. Targeting the PI3K/Akt/mTOR pathway may remove this inhibition and restore sensitivity to these compounds. Buparlisib (BKM120) is a potent oral pan-class I PI3K inhibitor that is being extensively evaluated in multiple tumor types. AREAS COVERED This review briefly summarizes the pharmacodynamics and pharmacokinetics of buparlisib, focusing on preclinical and clinical data in BC and on ongoing randomized trials. EXPERT OPINION Overall, buparlisib is a safe and tolerable drug and, despite its peculiar toxicity profile, it is suitable for studies in combination with other anticancer agents in BC. Early-phase clinical trials in BC have provided evidence of antitumor activity. Several trials are being conducted in all the biological subsets of BC, including combinations with endocrine therapy, anti-HER2 agents, PARP-inhibitors and chemotherapy. While clinical results are eagerly awaited, biological material suitable for both genomic and non-genomic analyses is being collected. The authors expect an intense investigation of the potential biomarkers that explain response or resistance to buparlisib and inspire strategies to rationally explore the therapeutic potential of this drug.
Collapse
Affiliation(s)
- Elena Geuna
- Investigative Clinical Oncology (INCO), Fondazione del Piemonte per l'Oncologia (FPO) - Candiolo Cancer Center (IRCCs), Institute for Cancer Research and Treatment of Candiolo , Strada Provinciale 142, Km 3.95, CAP 10060, Candiolo, Turin , Italy +39 0119933958 ; +39 0119621525 ;
| | | | | | | | | | | | | |
Collapse
|
99
|
Sestak I, Cuzick J. Markers for the identification of late breast cancer recurrence. Breast Cancer Res 2015. [PMID: 25848913 DOI: 10.1186/s13058-015-0156-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Postmenopausal women with early breast cancer are at an ongoing risk of relapse, even after successful surgery and treatment of the primary tumor. The treatment of breast cancer has changed in the past few years because of the discovery of prognostic and predictive biomarkers that allow individualized breast cancer treatment. However, it is still not clear how to identify women that are at high risk of a late recurrence. Clinical parameters are good prognostic markers for early recurrence, but only nodal status and, to a lesser extent, tumor size have proven to be strong prognostic markers for late recurrence. Multi-gene signatures have become widely used for the prediction of overall recurrence risk and tailoring administration of adjuvant chemotherapy, but only a few have been shown to be prognostic for late (distant) relapse. There is a need to accurately identify women who may benefit from extended endocrine therapy but also those who may be spared any additional treatment. Recent results from large clinical trials have shown that the research is going in the right direction, and these results might help to optimize extended endocrine therapy for patients with early breast cancer. However, further research is needed to select individual biomarkers or multi-gene signatures that offer identification of late recurrence specifically and thus justify routine use of these tests in the clinical setting.
Collapse
|
100
|
Abstract
Postmenopausal women with early breast cancer are at an ongoing risk of relapse, even after successful surgery and treatment of the primary tumor. The treatment of breast cancer has changed in the past few years because of the discovery of prognostic and predictive biomarkers that allow individualized breast cancer treatment. However, it is still not clear how to identify women that are at high risk of a late recurrence. Clinical parameters are good prognostic markers for early recurrence, but only nodal status and, to a lesser extent, tumor size have proven to be strong prognostic markers for late recurrence. Multi-gene signatures have become widely used for the prediction of overall recurrence risk and tailoring administration of adjuvant chemotherapy, but only a few have been shown to be prognostic for late (distant) relapse. There is a need to accurately identify women who may benefit from extended endocrine therapy but also those who may be spared any additional treatment. Recent results from large clinical trials have shown that the research is going in the right direction, and these results might help to optimize extended endocrine therapy for patients with early breast cancer. However, further research is needed to select individual biomarkers or multi-gene signatures that offer identification of late recurrence specifically and thus justify routine use of these tests in the clinical setting.
Collapse
Affiliation(s)
- Ivana Sestak
- Center for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| | - Jack Cuzick
- Center for Cancer Prevention, Wolfson Institute of Preventive Medicine, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ UK
| |
Collapse
|