1
|
Nakayama H, Murakami A, Nishida-Fukuda H, Fukuda S, Matsugi E, Nakahara M, Kusumoto C, Kamei Y, Higashiyama S. Semaphorin 3F inhibits breast cancer metastasis by regulating the Akt-mTOR and TGFβ signaling pathways via neuropilin-2. Sci Rep 2025; 15:7394. [PMID: 40033046 PMCID: PMC11876635 DOI: 10.1038/s41598-025-91559-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Class 3 semaphorins are axon guidance factors implicated in tumor and vascular biology, including invasive activity. Recent studies indicate that semaphorin 3F (SEMA3F) is a potent inhibitor of metastasis; however, its functional role in breast cancer is not fully understood. We found that exogenous SEMA3F inhibited phosphorylation of Akt and mTOR downstream kinase S6K in MDA-MB-231 and MCF7 cells via neuropilin-2 (NRP2) receptor. We also examined the effect of SEMA3F on breast cancer progression in vivo allograft model. The mouse 4T1 breast cancer cells or 4T1 cells overexpressing SEMA3F (4T1-SEMA3F) were implanted into mammary fat pads of Balb/c mice. We found that tumor growth was significantly inhibited in 4T1-SEMA3F injected mice compared to controls. Immunostaining revealed a remarkable reduction in the expression of vimentin, a mesenchymal cell marker, in 4T1-SEMA3F tumors. We also observed that mice injected with 4T1-SEMA3F cells had minimal metastasis to the liver and lungs, compared to controls. As a novel feature, SEMA3F suppressed TGFβ-induced Smad2 phosphorylation, resulting in the inhibition of cell invasiveness and epithelial-to-mesenchymal transition (EMT) in breast cancer. Consistently, a significant correlation between reduced expression of SEMA3F and poor outcome in patients with breast cancer. We conclude that SEMA3F acts as a dual inhibitor of the Akt-mTOR and TGFβ signaling pathways; thus, it has the potential to treat metastatic breast cancer.
Collapse
Affiliation(s)
- Hironao Nakayama
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan.
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, 791-0295, Ehime, Japan.
| | - Akari Murakami
- Breast Center, Ehime University Hospital, Toon, 791-0295, Ehime, Japan
| | - Hisayo Nishida-Fukuda
- Department of Biochemistry, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8650, Aichi, Japan
| | - Shinji Fukuda
- Department of Biochemistry, School of Dentistry, Aichi Gakuin University, Nagoya, 464-8650, Aichi, Japan
| | - Erina Matsugi
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Masako Nakahara
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Chiaki Kusumoto
- Department of Medical Science and Technology, Hiroshima International University, Higashi-hiroshima, 739-2695, Hiroshima, Japan
| | - Yoshiaki Kamei
- Breast Center, Ehime University Hospital, Toon, 791-0295, Ehime, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center (PROS), Ehime University, Toon, 791-0295, Ehime, Japan.
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Chuo-ku, Osaka, 541-8567, Japan.
| |
Collapse
|
2
|
Singh H. Role of Molecular Targeted Therapeutic Drugs in Treatment of Breast Cancer: A Review Article. Glob Med Genet 2023; 10:79-86. [PMID: 37228871 PMCID: PMC10205396 DOI: 10.1055/s-0043-57247] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023] Open
Abstract
Breast cancer is a multifactor, multistage, and heterogeneous disease. Systemic treatment of breast cancer has changed significantly over the last decade. With a better knowledge of the pathogenesis, researchers and scientists have discovered numerous signaling pathways and synonymous therapeutic targets in breast cancer. Because of the molecular nature of breast cancer, which makes it difficult to understand, previous attempts to treat or prevent it have failed. However, recent decades have provided effective therapeutic targets for treatment. In this review, literature or information on various targeted therapy for breast cancer is discussed. English language articles were explored in numerous directory or databases like PubMed, Web of Sciences, Google Scholar, ScienceDirect, and Scopus. The important keywords used for searching databases are "Breast cancer," "Targeted therapy in breast cancer," "Therapeutic drugs in breast cancer," and "Molecular targets in breast cancer."
Collapse
Affiliation(s)
- Himanshu Singh
- Department of Oral and Maxillofacial Pathology and Oral Microbiology, Index Institute of Dental Sciences, Indore, Madhya Pradesh, India
| |
Collapse
|
3
|
Pharmacological Inhibition of Lipid Import and Transport Proteins in Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14236004. [PMID: 36497485 PMCID: PMC9737127 DOI: 10.3390/cancers14236004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer (OC) is the most lethal gynecological malignancy with a 5-year survival rate of 49%. This is caused by late diagnosis when cells have already metastasized into the peritoneal cavity and to the omentum. OC progression is dependent on the availability of high-energy lipids/fatty acids (FA) provided by endogenous de novo biosynthesis and/or through import from the microenvironment. The blockade of these processes may thus represent powerful strategies against OC. While this has already been shown for inhibition of FA/lipid biosynthesis, evidence of the role of FA/lipid import/transport is still sparse. Therefore, we treated A2780 and SKOV3 OC cells with inhibitors of the lipid uptake proteins fatty acid translocase/cluster of differentiation 36 (FAT/CD36) and low-density lipoprotein (LDL) receptor (LDLR), as well as intracellular lipid transporters of the fatty acid-binding protein (FABP) family, fatty acid transport protein-2 (FATP2/SLC27A2), and ADP-ribosylation factor 6 (ARF6), which are overexpressed in OC. Proliferation was determined by formazan dye labeling/photometry and cell counting. Cell cycle analysis was performed by propidium iodide (PI) staining, and apoptosis was examined by annexin V/PI and active caspase 3 labeling and flow cytometry. RNA-seq data revealed altered stress and metabolism pathways. Overall, the small molecule inhibitors of lipid handling proteins BMS309403, HTS01037, NAV2729, SB-FI-26, and sulfosuccinimidyl oleate (SSO) caused a drug-specific, dose-/time-dependent inhibition of FA/LDL uptake, associated with reduced proliferation, cell cycle arrest, and apoptosis. Our findings indicate that OC cells are very sensitive to lipid deficiency. This dependency should be exploited for development of novel strategies against OC.
Collapse
|
4
|
Gámez-Chiachio M, Sarrió D, Moreno-Bueno G. Novel Therapies and Strategies to Overcome Resistance to Anti-HER2-Targeted Drugs. Cancers (Basel) 2022; 14:4543. [PMID: 36139701 PMCID: PMC9496705 DOI: 10.3390/cancers14184543] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/09/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
The prognosis and quality of life of HER2 breast cancer patients have significantly improved due to the crucial clinical benefit of various anti-HER2 targeted therapies. However, HER2 tumors can possess or develop several resistance mechanisms to these treatments, thus leaving patients with a limited set of additional therapeutic options. Fortunately, to overcome this problem, in recent years, multiple different and complementary approaches have been developed (such as antibody-drug conjugates (ADCs)) that are in clinical or preclinical stages. In this review, we focus on emerging strategies other than on ADCs that are either aimed at directly target the HER2 receptor (i.e., novel tyrosine kinase inhibitors) or subsequent intracellular signaling (e.g., PI3K/AKT/mTOR, CDK4/6 inhibitors, etc.), as well as on innovative approaches designed to attack other potential tumor weaknesses (such as immunotherapy, autophagy blockade, or targeting of other genes within the HER2 amplicon). Moreover, relevant technical advances such as anti-HER2 nanotherapies and immunotoxins are also discussed. In brief, this review summarizes the impact of novel therapeutic approaches on current and future clinical management of aggressive HER2 breast tumors.
Collapse
Affiliation(s)
- Manuel Gámez-Chiachio
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - David Sarrió
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Gema Moreno-Bueno
- Biochemistry Department, Medicine Faculty, Universidad Autónoma Madrid-CSIC, IdiPaz, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
- MD Anderson International Foundation, 28033 Madrid, Spain
| |
Collapse
|
5
|
Dunn E, Chitcholtan K, Sykes P, Garrill A. The Anti-Proliferative Effect of PI3K/mTOR and ERK Inhibition in Monolayer and Three-Dimensional Ovarian Cancer Cell Models. Cancers (Basel) 2022; 14:cancers14020395. [PMID: 35053555 PMCID: PMC8773481 DOI: 10.3390/cancers14020395] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In ovarian cancer patients the PI3K/AKT/mTOR and RAS/RAF/MEK/ERK kinase signaling pathways are frequently dysregulated, making them potential targets of therapeutic inhibitors. In this study, we used four human ovarian cancer cell lines grown in two- and three-dimensional models to investigate the potential efficacy of combining two inhibitors, which target these pathways, against ovarian cancer. The inhibitor combination was found to have cell line- and model-dependent synergistic antiproliferative effect. Abstract Most ovarian cancer patients are diagnosed with advanced stage disease, which becomes unresponsive to chemotherapeutic treatments. The PI3K/AKT/mTOR and the RAS/RAF/MEK/ERK kinase signaling pathways are attractive targets for potential therapeutic inhibitors, due to the high frequency of mutations to PTEN, PIK3CA, KRAS and BRAF in several ovarian cancer subtypes. However, monotherapies targeting one of these pathways have shown modest effects in clinical trials. This limited efficacy of the agents could be due to upregulation and increased signaling via the adjacent alternative pathway. In this study, the efficacy of combined PI3K/mTOR (BEZ235) and ERK inhibition (SCH772984) was investigated in four human ovarian cancer cell lines, grown as monolayer and three-dimensional cell aggregates. The inhibitor combination reduced cellular proliferation in a synergistic manner in OV-90 and OVCAR8 monolayers and in OV-90, OVCAR5 and SKOV3 aggregates. Sensitivity to the inhibitors was reduced in three-dimensional cell aggregates in comparison to monolayers. OV-90 cells cultured in large spheroids were sensitive to the inhibitors and displayed a robust synergistic antiproliferative response to the inhibitor combination. In contrast, OVCAR8 spheroids were resistant to the inhibitors. These findings suggest that combined PI3K/mTOR and ERK inhibition could be a useful strategy for overcoming treatment resistance in ovarian cancer and warrants further preclinical investigation. Additionally, in some cell lines the use of different three-dimensional models can influence cell line sensitivity to PI3K/mTOR and RAS/RAF/MEK/ERK pathway inhibitors.
Collapse
Affiliation(s)
- Elizabeth Dunn
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
- Correspondence: (E.D.); (A.G.)
| | - Kenny Chitcholtan
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch 8011, New Zealand; (K.C.); (P.S.)
| | - Peter Sykes
- Department of Obstetrics and Gynaecology, University of Otago, Christchurch 8011, New Zealand; (K.C.); (P.S.)
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
- Biomolecular Interaction Centre, School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
- Correspondence: (E.D.); (A.G.)
| |
Collapse
|
6
|
Sanz-Álvarez M, Martín-Aparicio E, Luque M, Zazo S, Martínez-Useros J, Eroles P, Rovira A, Albanell J, Madoz-Gúrpide J, Rojo F. The Novel Oral mTORC1/2 Inhibitor TAK-228 Reverses Trastuzumab Resistance in HER2-Positive Breast Cancer Models. Cancers (Basel) 2021; 13:cancers13112778. [PMID: 34204960 PMCID: PMC8199905 DOI: 10.3390/cancers13112778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Hyperactivation of the PI3K/AKT/mTOR cell signalling pathway is an important and well-described mechanism of trastuzumab resistance in HER2-positive breast cancer. In cell-line models of acquired trastuzumab resistance generated in our laboratory, we demonstrate this type of activation, which is independent of HER2-mediated regulation. We investigate whether the use of specific mTOR inhibitors, a PI3K/AKT/mTOR pathway effector, could lead to decreased activity of the pathway, influencing trastuzumab resistance. We demonstrate that TAK-228, a mTORC1 and mTORC2 inhibitor, can reverse resistance and increasing response to trastuzumab in models of primary and acquired resistance. Abstract The use of anti-HER2 therapies has significantly improved clinical outcome in patients with HER2-positive breast cancer, yet a substantial proportion of patients acquire resistance after a period of treatment. The PI3K/AKT/mTOR pathway is a good target for drug development, due to its involvement in HER2-mediated signalling and in the emergence of resistance to anti-HER2 therapies, such as trastuzumab. This study evaluates the activity of three different PI3K/AKT/mTOR inhibitors, i.e., BEZ235, everolimus and TAK-228 in vitro, in a panel of HER2-positive breast cancer cell lines with primary and acquired resistance to trastuzumab. We assess the antiproliferative effect and PI3K/AKT/mTOR inhibitory capability of BEZ235, everolimus and TAK-228 alone, and in combination with trastuzumab. Dual blockade with trastuzumab and TAK-228 was superior in reversing the acquired resistance in all the cell lines. Subsequently, we analyse the effects of TAK-228 in combination with trastuzumab on the cell cycle and found a significant increase in G0/G1 arrest in most cell lines. Likewise, the combination of both drugs induced a significant increase in apoptosis. Collectively, these experiments support the combination of trastuzumab with PI3K/AKT/mTOR inhibitors as a potential strategy for inhibiting the proliferation of HER2-positive breast cancer cell lines that show resistance to trastuzumab.
Collapse
Affiliation(s)
- Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Ester Martín-Aparicio
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Melani Luque
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Sandra Zazo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain;
| | - Pilar Eroles
- Institute of Health Research INCLIVA-CIBERONC, 46010 Valencia, Spain;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Faculty of Medicine, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| |
Collapse
|
7
|
Grunt TW, Lemberger L, Colomer R, López Rodríguez ML, Wagner R. The Pharmacological or Genetic Blockade of Endogenous De Novo Fatty Acid Synthesis Does Not Increase the Uptake of Exogenous Lipids in Ovarian Cancer Cells. Front Oncol 2021; 11:610885. [PMID: 33928023 PMCID: PMC8076863 DOI: 10.3389/fonc.2021.610885] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 03/19/2021] [Indexed: 12/28/2022] Open
Abstract
Ovarian cancer(OC) is a serious threat to women worldwide. Peritoneal dissemination, ascites and omental metastasis are typical features for disease progression, which occurs in a micro-environment that is rich in high-energy lipids. OC cells require high amounts of lipids for survival and growth. Not only do they import lipids from the host, they also produce lipids de novo. Inhibitors of fatty acid(FA) synthase(FASN) – the rate-limiting enzyme of endogenous FA synthesis that is overexpressed in OC – induce growth-arrest and apoptosis, rendering them promising candidates for cancer drug development. However, cancer researchers have long hypothesized that the lipid deficiency caused by FASN inhibition can be circumvented by increasing the uptake of exogenous lipids from the host, which would confer resistance to FASN inhibitors. In contrast to a very recent report in colorectal cancer, we demonstrate in OC cells (A2780, OVCAR3, SKOV3) that neither FASN inhibitors (G28UCM, Fasnall) nor FASN-specific siRNAs can stimulate a relief pathway leading to enhanced uptake of extrinsic FAs or low density lipoproteins (LDLs). Instead, we observed that the growth-arrest due to FASN inhibition or FASN knock-down was associated with significant dose- and time-dependent reduction in the uptake of fluorescently labeled FAs and LDLs. Western blotting showed that the expression of the FA receptor CD36, the LDL receptor(LDLR) and the lipid transport proteins fatty acid binding proteins 1–9 (FABP1–9) was not affected by the treatment. Next, we compared experimental blockade of endogenous lipid production with physiologic depletion of exogenous lipids. Lipid-free media, similar to FASN inhibitors, caused growth-arrest. Although lipid-depleted cells have diminished amounts of CD36, LDLR and FABPs, they can still activate a restorative pathway that causes enhanced import of fluorophore-labeled FAs and LDLs. Overall, our data show that OC cells are strictly lipid-depend and exquisitely sensitive to FASN inhibitors, providing a strong rationale for developing anti-FASN strategies for clinical use against OC.
Collapse
Affiliation(s)
- Thomas W Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Comprehensive Cancer Center, Vienna, Austria.,Ludwig Boltzmann Institute for Hematology and Oncology, Vienna, Austria
| | - Lisa Lemberger
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ramón Colomer
- Clinical Research Program, Department of Medical Oncology, Hospital Universitario La Princesa and Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María Luz López Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Renate Wagner
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria.,Comprehensive Cancer Center, Vienna, Austria
| |
Collapse
|
8
|
Dong C, Wu J, Chen Y, Nie J, Chen C. Activation of PI3K/AKT/mTOR Pathway Causes Drug Resistance in Breast Cancer. Front Pharmacol 2021; 12:628690. [PMID: 33790792 PMCID: PMC8005514 DOI: 10.3389/fphar.2021.628690] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Although chemotherapy, targeted therapy and endocrine therapy decrease rate of disease recurrence in most breast cancer patients, many patients exhibit acquired resistance. Hyperactivation of the PI3K/AKT/mTOR pathway is associated with drug resistance and cancer progression. Currently, a number of drugs targeting PI3K/AKT/mTOR are being investigated in clinical trials by combining them with standard therapies to overcome acquired resistance in breast cancer. In this review, we summarize the critical role of the PI3K/AKT/mTOR pathway in drug resistance, the development of PI3K/AKT/mTOR inhibitors, and strategies to overcome acquired resistance to standard therapies in breast cancer.
Collapse
Affiliation(s)
- Chao Dong
- Department of the Second Medical Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Jiao Wu
- Department of the Second Medical Oncology, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Yin Chen
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai, China
| | - Jianyun Nie
- Department of the Third Breast Surgery, The 3rd Affiliated Hospital of Kunming Medical University, Yunnan Tumor Hospital, Kunming, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
9
|
Grunt TW, Slany A, Semkova M, Colomer R, López-Rodríguez ML, Wuczkowski M, Wagner R, Gerner C, Stübiger G. Membrane disruption, but not metabolic rewiring, is the key mechanism of anticancer-action of FASN-inhibitors: a multi-omics analysis in ovarian cancer. Sci Rep 2020; 10:14877. [PMID: 32913236 PMCID: PMC7483762 DOI: 10.1038/s41598-020-71491-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 07/15/2020] [Indexed: 01/12/2023] Open
Abstract
Fatty-acid(FA)-synthase(FASN) is a druggable lipogenic oncoprotein whose blockade causes metabolic disruption. Whether drug-induced metabolic perturbation is essential for anticancer drug-action, or is just a secondary—maybe even a defence response—is still unclear. To address this, SKOV3 and OVCAR3 ovarian cancer(OC) cell lines with clear cell and serous histology, two main OC subtypes, were exposed to FASN-inhibitor G28UCM. Growth-inhibition was compared with treatment-induced cell-metabolomes, lipidomes, proteomes and kinomes. SKOV3 and OVCAR3 were equally sensitive to low-dose G28UCM, but SKOV3 was more resistant than OVCAR3 to higher concentrations. Metabolite levels generally decreased upon treatment, but individual acylcarnitines, glycerophospholipids, sphingolipids, amino-acids, biogenic amines, and monosaccharides reacted differently. Drug-induced effects on central-carbon-metabolism and oxidative-phosphorylation (OXPHOS) were essentially different in the two cell lines, since drug-naïve SKOV3 are known to prefer glycolysis, while OVCAR3 favour OXPHOS. Moreover, drug-dependent increase of desaturases and polyunsaturated-fatty-acids (PUFAs) were more pronounced in SKOV3 and appear to correlate with G28UCM-tolerance. In contrast, expression and phosphorylation of proteins that control apoptosis, FA synthesis and membrane-related processes (beta-oxidation, membrane-maintenance, transport, translation, signalling and stress-response) were concordantly affected. Overall, membrane-disruption and second-messenger-silencing were crucial for anticancer drug-action, while metabolic-rewiring was only secondary and may support high-dose-FASN-inhibitor-tolerance. These findings may guide future anti-metabolic cancer intervention.
Collapse
Affiliation(s)
- Thomas W Grunt
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria. .,Comprehensive Cancer Center, Vienna, Austria. .,Ludwig Boltzmann Institute for Hematology and Oncology, Vienna, Austria.
| | - Astrid Slany
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Mariya Semkova
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Ramón Colomer
- Department of Medical Oncology, Hospital Universitario La Princesa and Spanish National Cancer Research Centre (CNIO), Clinical Research Program, Madrid, Spain
| | - María Luz López-Rodríguez
- Departamento de Química Orgánica I, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Michael Wuczkowski
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Renate Wagner
- Cell Signaling and Metabolism Networks Program, Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.,Comprehensive Cancer Center, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Gerald Stübiger
- Comprehensive Cancer Center, Vienna, Austria.,Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
10
|
Maldonado MDM, Medina JI, Velazquez L, Dharmawardhane S. Targeting Rac and Cdc42 GEFs in Metastatic Cancer. Front Cell Dev Biol 2020; 8:201. [PMID: 32322580 PMCID: PMC7156542 DOI: 10.3389/fcell.2020.00201] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
The Rho family GTPases Rho, Rac, and Cdc42 have emerged as key players in cancer metastasis, due to their essential roles in regulating cell division and actin cytoskeletal rearrangements; and thus, cell growth, migration/invasion, polarity, and adhesion. This review will focus on the close homologs Rac and Cdc42, which have been established as drivers of metastasis and therapy resistance in multiple cancer types. Rac and Cdc42 are often dysregulated in cancer due to hyperactivation by guanine nucleotide exchange factors (GEFs), belonging to both the diffuse B-cell lymphoma (Dbl) and dedicator of cytokinesis (DOCK) families. Rac/Cdc42 GEFs are activated by a myriad of oncogenic cell surface receptors, such as growth factor receptors, G-protein coupled receptors, cytokine receptors, and integrins; consequently, a number of Rac/Cdc42 GEFs have been implicated in metastatic cancer. Hence, inhibiting GEF-mediated Rac/Cdc42 activation represents a promising strategy for targeted metastatic cancer therapy. Herein, we focus on the role of oncogenic Rac/Cdc42 GEFs and discuss the recent advancements in the development of Rac and Cdc42 GEF-interacting inhibitors as targeted therapy for metastatic cancer, as well as their potential for overcoming cancer therapy resistance.
Collapse
Affiliation(s)
- Maria Del Mar Maldonado
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Julia Isabel Medina
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Luis Velazquez
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| | - Suranganie Dharmawardhane
- Department of Biochemistry, School of Medicine, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico
| |
Collapse
|
11
|
Feng WW, Kurokawa M. Lipid metabolic reprogramming as an emerging mechanism of resistance to kinase inhibitors in breast cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3. [PMID: 32226926 PMCID: PMC7100881 DOI: 10.20517/cdr.2019.100] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Breast cancer is one of the leading causes of death in women in the United States. In general, patients with breast cancer undergo surgical resection of the tumor and/or receive drug treatment to kill or suppress the growth of cancer cells. In this regard, small molecule kinase inhibitors serve as an important class of drugs used in clinical and research settings. However, the development of resistance to these compounds, in particular HER2 and CDK4/6 inhibitors, often limits durable clinical responses to therapy. Emerging evidence indicates that PI3K/AKT/mTOR pathway hyperactivation is one of the most prominent mechanisms of resistance to many small molecule inhibitors as it bypasses upstream growth factor receptor inhibition. Importantly, the PI3K/AKT/mTOR pathway also plays a pertinent role in regulating various aspects of cancer metabolism. Recent studies from our lab and others have demonstrated that altered lipid metabolism mediates the development of acquired drug resistance to HER2-targeted therapies in breast cancer, raising an interesting link between reprogrammed kinase signaling and lipid metabolism. It appears that, upon development of resistance to HER2 inhibitors, breast cancer cells rewire lipid metabolism to somehow circumvent the inhibition of kinase signaling. Here, we review various mechanisms of resistance observed for kinase inhibitors and discuss lipid metabolism as a potential therapeutic target to overcome acquired drug resistance.
Collapse
Affiliation(s)
- William W Feng
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Manabu Kurokawa
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA.,Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| |
Collapse
|
12
|
McKenna M, McGarrigle S, Pidgeon GP. The next generation of PI3K-Akt-mTOR pathway inhibitors in breast cancer cohorts. Biochim Biophys Acta Rev Cancer 2018; 1870:185-197. [PMID: 30318472 DOI: 10.1016/j.bbcan.2018.08.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/09/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
The PI3K/Akt/mTOR pathway plays a role in various oncogenic processes in breast cancer and key pathway aberrations have been identified which drive the different molecular subtypes. Early drugs developed targeting this pathway produced some clinical success but were hampered by pharmacokinetics, tolerability and efficacy problems. This created a need for new PI3K pathway-inhibiting drugs, which would produce more robust results allowing incorporation into treatment regimens for breast cancer patients. In this review, the most promising candidates from the new generation of PI3K-pathway inhibitors is explored, presenting evidence from preclinical and early clinical research, as well as ongoing trials utilising these drugs in breast cancer cohorts. The problems hindering the development of drugs targeting the PI3K pathway are examined, which have created problems for their use as monotherapies. PI3K pathway inhibitor combinations therefore remains a dynamic research area, and their role in combination with immunotherapies and epigenetic therapies is also inspected.
Collapse
Affiliation(s)
- Michael McKenna
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Sarah McGarrigle
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
| | - Graham P Pidgeon
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
13
|
|
14
|
Abstract
The efficient production, folding, and secretion of proteins is critical for cancer cell survival. However, cancer cells thrive under stress conditions that damage proteins, so many cancer cells overexpress molecular chaperones that facilitate protein folding and target misfolded proteins for degradation via the ubiquitin-proteasome or autophagy pathway. Stress response pathway induction is also important for cancer cell survival. Indeed, validated targets for anti-cancer treatments include molecular chaperones, components of the unfolded protein response, the ubiquitin-proteasome system, and autophagy. We will focus on links between breast cancer and these processes, as well as the development of drug resistance, relapse, and treatment.
Collapse
Affiliation(s)
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, 4249 Fifth Ave, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
15
|
Gschwantler-Kaulich D, Weingartshofer S, Grunt TW, Mairhofer M, Tan Y, Gamper J, Singer CF. Estradiol impairs the antiproliferative and proapoptotic effect of Zoledronic acid in hormone sensitive breast cancer cells in vitro. PLoS One 2017; 12:e0185566. [PMID: 28945801 PMCID: PMC5612728 DOI: 10.1371/journal.pone.0185566] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/14/2017] [Indexed: 11/19/2022] Open
Abstract
Background Zoledronic acid (ZA) has antiresorptive effects and protects from bone metastasis in women with early breast cancer. In addition, in postmenopausal women with endocrine responsive breast cancer ZA prolongs DFS. The exact mechanism is still unclear. We have therefore investigated the effect of increasing concentrations of ZA in breast cancer cell lines in the absence or presence of estradiol to mimic the hormonal environment in vitro. Materials and methods Using assays for cell proliferation (EZ4U, BrdU) and cell death (Annexin/PI), we have analyzed the dose-dependent antiproliferative and pro-apoptotic effects of ZA in two hormone sensitive cell lines (MCF-7 and T47D) and a hormone insensitive, triple negative cell line (MDA-MB-231) in the presence of 0, 1 and 10 nM estradiol. Results In the absence of estradiol, ZA exerts dose-dependent antiproliferative and pro-apoptotic antitumor effects in both, hormone sensitive (MCF-7, T47D) and -insensitive (MDA-MB-231) breast cancer cell lines (p<0.0001). In the presence of estradiol, the antitumoral effect of ZA was significantly decreased only in the hormone sensitive MCF-7 and T47D cell lines (p = 0.0008 and p = 0.0008, respectively). Conclusion We have demonstrated that estradiol impairs the antiproliferative and proapoptotic effect of ZA in hormone sensitive, but not in hormone insensitive breast cancer cell lines. Our findings provide a possible explanation for the differential effect of ZA on DFS in pre- and postmenopausal patients with hormone sensitive early breast cancer, which has been demonstrated clinically. We further hypothesize that endocrine insensitive tumors such as triple negative breast cancer (TNBC) should benefit from ZA irrespective of their menopausal status.
Collapse
Affiliation(s)
- Daphne Gschwantler-Kaulich
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- * E-mail:
| | - Sigrid Weingartshofer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Thomas W. Grunt
- Clinical Division of Oncology, Department of Medicine I, Comprehensive Cancer Center & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Vienna, Austria
| | | | - Yen Tan
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- QIMR Berghofer Medical Research Institute, Herston QLD, Australia
| | - Jutta Gamper
- Section for Medical Statistics, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Christian F. Singer
- Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
16
|
Guerin M, Rezai K, Isambert N, Campone M, Autret A, Pakradouni J, Provansal M, Camerlo J, Sabatier R, Bertucci F, Charafe-Jauffret E, Hervieu A, Extra JM, Viens P, Lokiec F, Boher JM, Gonçalves A. PIKHER2: A phase IB study evaluating buparlisib in combination with lapatinib in trastuzumab-resistant HER2-positive advanced breast cancer. Eur J Cancer 2017; 86:28-36. [PMID: 28950146 DOI: 10.1016/j.ejca.2017.08.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 10/18/2022]
Abstract
BACKGROUND Phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin pathway is frequently activated in HER2-positive breast cancer and may play a major role in resistance to trastuzumab. Buparlisib is a pan-class-I PI3K inhibitor with potent and selective activity against wild-type and mutant PI3K p110 isoforms. PATIENTS AND METHODS PIKHER2 phase IB study aimed primarily to determine a maximum tolerated dose (MTD) and propose a recommended phase II dose (RP2D) for buparlisib in combination with lapatinib in HER2-positive, trastuzumab-resistant, advanced breast cancer. Oral buparlisib (40, 60 or 80 mg) and lapatinib (750, 1000 or 1250 mg) were administered daily. A modified continuous reassessment method using an adaptive Bayesian model guided the dose escalation of both agents. Secondary end-points included antitumour activity and pharmacokinetic (PK) assessments. RESULTS A total of 24 patients were treated across five dose levels. Dose-limiting toxicities included transaminases elevation, vomiting, stomatitis, hyperglycemia and diarrhoea. MTD was declared at buparlisib 80 mg/d + lapatinib 1250 mg/d, but toxicities and early treatment discontinuation rate beyond cycle 1 led to select buparlisib 80 mg + lapatinib 1000 mg/d as the RP2D. Main drug-related adverse events included diarrhoea, nausea, skin rash, asthenia, depression, anxiety and transaminases increase. There was no significant evidence for drug-drug PK interaction. Disease control rate was 79% [95% confidence interval [CI] 57-92%], one patient obtained a complete remission, and six additional patients experienced stable disease for ≥ 24 weeks (clinical benefit rate of 29% [95% CI 12-51%]). CONCLUSION Combining buparlisib and lapatinib in HER2-positive trastuzumab-resistant advanced breast cancer was feasible. Preliminary evidence of antitumour activity was observed in this heavily pre-treated population. TRIAL REGISTRATION ID NCT01589861.
Collapse
Affiliation(s)
- Mathilde Guerin
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Keyvan Rezai
- Institut Curie - Hôpital René Huguenin, Saint-Cloud, France
| | | | | | - Aurélie Autret
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France
| | - Jihane Pakradouni
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France
| | - Magali Provansal
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Jacques Camerlo
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Renaud Sabatier
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - François Bertucci
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Emmanuelle Charafe-Jauffret
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Biopathology, CRCM, Marseille, France
| | | | - Jean-Marc Extra
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | - Patrice Viens
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France
| | | | - Jean-Marie Boher
- Institut Paoli-Calmettes, Department of Clinical Research and Innovations, Marseille, France; Aix Marseille University, INSERM, IRD, SESSTIM, Sciences Economiques & Sociales de La Santé & Traitement de L'Information Médicale, Marseille, France
| | - Anthony Gonçalves
- Aix-Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, Department of Medical Oncology, CRCM, Marseille, France.
| |
Collapse
|
17
|
Li Y, Zhang X, Niu S, Zhao Y, Yang L, Shao X, Wang E. Synthesis and biological activity of imidazo[4,5-c]quinoline derivatives as PI3K/mTOR inhibitors. Chem Res Chin Univ 2017. [DOI: 10.1007/s40242-017-7074-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Multi-level suppression of receptor-PI3K-mTORC1 by fatty acid synthase inhibitors is crucial for their efficacy against ovarian cancer cells. Oncotarget 2017; 8:11600-11613. [PMID: 28086243 PMCID: PMC5355289 DOI: 10.18632/oncotarget.14591] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 12/24/2016] [Indexed: 01/19/2023] Open
Abstract
Receptor-PI3K-mTORC1 signaling and fatty acid synthase (FASN)-regulated lipid biosynthesis harbor numerous drug targets and are molecularly connected. We hypothesize that unraveling the mechanisms of pathway cross-talk will be useful for designing novel co-targeting strategies for ovarian cancer (OC). The impact of receptor-PI3K-mTORC1 onto FASN is already well-characterized. However, reverse actions–from FASN towards receptor-PI3K-mTORC1–are still elusive. We show that FASN-blockade impairs receptor-PI3K-mTORC1 signaling at multiple levels. Thin-layer chromatography and MALDI-MS/MS reveals that FASN-inhibitors (C75, G28UCM) augment polyunsaturated fatty acids and diminish signaling lipids diacylglycerol (DAG) and phosphatidylinositol 3,4,5-trisphosphate (PIP3) in OC cells (SKOV3, OVCAR-3, A2780, HOC-7). Western blotting and micropatterning demonstrate that FASN-blockers impair phosphorylation/expression of EGF-receptor/ERBB/HER and decrease GRB2–EGF-receptor recruitment leading to PI3K-AKT suppression. FASN-inhibitors activate stress response-genes HIF-1α-REDD1 (RTP801/DIG2/DDIT4) and AMPKα causing mTORC1- and S6-repression. We conclude that FASN-inhibitor-mediated blockade of receptor-PI3K-mTORC1 occurs due to a number of distinct but cooperating processes. Moreover, decrease of PI3K-mTORC1 abolishes cross-repression of MEK-ERK causing ERK activation. Consequently, the MEK-inhibitor selumetinib/AZD6244, in contrast to the PI3K/mTOR-inhibitor dactolisib/NVP-BEZ235, increases growth inhibition when given together with a FASN-blocker. We are the first to provide deep insight on how FASN-inhibition blocks ERBB-PI3K-mTORC1 activity at multiple molecular levels. Moreover, our data encourage therapeutic approaches using FASN-antagonists together with MEK-ERK-inhibitors.
Collapse
|
19
|
Corona SP, Ravelli A, Cretella D, Cappelletti MR, Zanotti L, Dester M, Gobbi A, Petronini PG, Generali D. CDK4/6 inhibitors in HER2-positive breast cancer. Crit Rev Oncol Hematol 2017; 112:208-214. [PMID: 28325261 DOI: 10.1016/j.critrevonc.2017.02.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/20/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022] Open
Abstract
Notwithstanding the continuous progress made in cancer treatment in the last 20 years, and the availability of new targeted therapies, metastatic Breast Cancer (BC) is still incurable. Targeting the cell cycle machinery has emerged as an attractive strategy to tackle cancer progression, showing very promising results in the preclinical and clinical settings. The first selective inhibitors of CDK4/6 received breakthrough status and FDA approval in combination with letrozole (February 2015) and fulvestrant (February 2016) as first-line therapy in ER-positive advanced and metastatic BC. Considering the success of this family of compounds in hormone-positive BC, new possible applications are being investigated in other molecular subtypes. This review summarizes the latest findings on the use of CDK4/6 inhibitors in HER2 positive BC.
Collapse
Affiliation(s)
- Silvia Paola Corona
- Peter MacCallum Cancer Centre, Radiation Oncology Department, Moorabbin Campus, East Bentleigh Victoria 3165, Australia.
| | - Andrea Ravelli
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Daniele Cretella
- Universita degli Studi di Parma, Department of Clinical and Experimental Medicine, Experimental Oncology Unit, Via Gramsci, 14, Parma, Italy
| | - Maria Rosa Cappelletti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Laura Zanotti
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Martina Dester
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Angela Gobbi
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Pier Giorgio Petronini
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy
| | - Daniele Generali
- Azienda Ospedaliera di Cremona, U.O. Multidisciplinare di Patologia Mammaria, U.S. Terapia Molecolare e Farmacogenomica, Cremona, Italy; Universita degli Studi di Trieste, Department of Medical, Surgery and Health Sciences, Trieste, Italy
| |
Collapse
|
20
|
Deblois G, Smith HW, Tam IS, Gravel SP, Caron M, Savage P, Labbé DP, Bégin LR, Tremblay ML, Park M, Bourque G, St-Pierre J, Muller WJ, Giguère V. ERRα mediates metabolic adaptations driving lapatinib resistance in breast cancer. Nat Commun 2016; 7:12156. [PMID: 27402251 PMCID: PMC4945959 DOI: 10.1038/ncomms12156] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 06/06/2016] [Indexed: 12/27/2022] Open
Abstract
Despite the initial benefits of treating HER2-amplified breast cancer patients with the tyrosine kinase inhibitor lapatinib, resistance inevitably develops. Here we report that lapatinib induces the degradation of the nuclear receptor ERRα, a master regulator of cellular metabolism, and that the expression of ERRα is restored in lapatinib-resistant breast cancer cells through reactivation of mTOR signalling. Re-expression of ERRα in resistant cells triggers metabolic adaptations favouring mitochondrial energy metabolism through increased glutamine metabolism, as well as ROS detoxification required for cell survival under therapeutic stress conditions. An ERRα inverse agonist counteracts these metabolic adaptations and overcomes lapatinib resistance in a HER2-induced mammary tumour mouse model. This work reveals a molecular mechanism by which ERRα-induced metabolic reprogramming promotes survival of lapatinib-resistant cancer cells and demonstrates the potential of ERRα inhibition as an effective adjuvant therapy in poor outcome HER2-positive breast cancer.
Collapse
Affiliation(s)
- Geneviève Deblois
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Harvey W. Smith
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Ingrid S. Tam
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Simon-Pierre Gravel
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Maxime Caron
- Department of Human Genetics, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Paul Savage
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - David P. Labbé
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Louis R. Bégin
- Service d'anatomopathologie, Hôpital du Sacré-Cœur de Montréal, 5400 Boulevard Gouin Ouest, Montréal, Québec, Canada H4J 1C5
| | - Michel L. Tremblay
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - Julie St-Pierre
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
| | - William J. Muller
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Biochemistry, McGill University, Montréal, Québec, Canada H3G 1Y6
- Department of Medicine, McGill University, Montréal, Québec, Canada H3A 1A3
- Department of Oncology, McGill University, Montréal, Québec, Canada H2W 1S6
| |
Collapse
|
21
|
HER Specific TKIs Exert Their Antineoplastic Effects on Breast Cancer Cell Lines through the Involvement of STAT5 and JNK. PLoS One 2016; 11:e0146311. [PMID: 26735495 PMCID: PMC4703392 DOI: 10.1371/journal.pone.0146311] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/15/2015] [Indexed: 12/14/2022] Open
Abstract
Background HER-targeted tyrosine kinase inhibitors (TKIs) have demonstrated pro-apoptotic and antiproliferative effects in vitro and in vivo. The exact pathways through which TKIs exert their antineoplastic effects are, however, still not completely understood. Methods Using Milliplex assays, we have investigated the effects of the three panHER-TKIs lapatinib, canertinib and afatinib on signal transduction cascade activation in SKBR3, T47D and Jurkat neoplastic cell lines. The growth-inhibitory effect of blockade of HER and of JNK and STAT5 signaling was measured by proliferation- and apoptosis-assays using formazan dye labeling of viable cells, Western blotting for cleaved PARP-1 and immunolabeling for active caspase 3, respectively. Results All three HER-TKIs clearly inhibited proliferation and increased apoptosis in HER2 overexpressing SKBR3 cells, while their effect was less pronounced on HER2 moderately expressing T47D cells where they exerted only a weak antiproliferative and essentially no pro-apoptotic effect. Remarkably, phosphorylation/activation of JNK and STAT5A/B were inhibited by HER-TKIs only in the sensitive, but not in the resistant cells. In contrast, phosphorylation/activation of ERK/MAPK, STAT3, CREB, p70 S6 kinase, IkBa, and p38 were equally affected by HER-TKIs in both cell lines. Moreover, we demonstrated that direct pharmacological blockade of JNK and STAT5 abrogates cell growth in both HER-TKI-sensitive as well as -resistant breast cancer cells, respectively. Conclusion We have shown that HER-TKIs exert a HER2 expression-dependent anti-cancer effect in breast cancer cell lines. This involves blockade of JNK and STAT5A/B signaling, which have been found to be required for in vitro growth of these cell lines.
Collapse
|
22
|
Fazio N. Neuroendocrine tumors resistant to mammalian target of rapamycin inhibitors: A difficult conversion from biology to the clinic. World J Clin Oncol 2015; 6:194-197. [PMID: 26677429 PMCID: PMC4675901 DOI: 10.5306/wjco.v6.i6.194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/07/2015] [Accepted: 09/25/2015] [Indexed: 02/06/2023] Open
Abstract
Deregulation of the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) - mammalian target of rapamycin (mTOR) signaling pathway is one of the most commonly-involved pathways in tumorigenesis. It has also been reported as altered in neuroendocrine tumors (NETs). mTOR inhibitors used in clinical practice are derived from rapamycin, an anti-cancer agent also used as an immunosuppressor after organ transplantation. Everolimus and temsirolimus are the two rapamycin-derived mTOR inhibitors used in NETs. Notably everolimus has been approved in advanced progressive well/moderately-differentiated pancreatic NETs (pNETs). It inhibits specifically the mTORC1 subunit of mTOR, not interacting with mTORC2. Although everolimus produced a significant prolongation of progression-free survival a number of patients with pNETs do not benefit from the drug due to early or late progression. Two supposed mechanisms of resistance to mTOR inhibitors are Akt and PI3K activation, by means of mTORC2 and insulin growth factor (IGF) - IGF receptor signaling, respectively. BEZ235 is a multi-targeted inhibitor binding to PI3K, mTORC1 and mTORC2, therefore potentially turning off all the supposed molecular targets of resistance to everolimus. The two clinical trials designed in pNETs were stopped early due to unmet statistical endpoint and the global clinical development of BEZ235 was also halted. Tolerability of this drug was challenging and conditioned the feasibility of therapy. The BEZ experience is an example of the huge difference between the preclinical and clinical setting and prompts us to pay more attention to the phase I step of clinical development and the design of phase II clinical trials.
Collapse
|
23
|
Lee JJ, Loh K, Yap YS. PI3K/Akt/mTOR inhibitors in breast cancer. Cancer Biol Med 2015; 12:342-54. [PMID: 26779371 PMCID: PMC4706528 DOI: 10.7497/j.issn.2095-3941.2015.0089] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 11/18/2015] [Indexed: 12/17/2022] Open
Abstract
Activation of the phosphoinositide 3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway is common in breast cancer. There is preclinical data to support inhibition of the pathway, and phase I to III trials involving inhibitors of the pathway have been or are being conducted in solid tumors and breast cancer. Everolimus, an mTOR inhibitor, is currently approved for the treatment of hormone receptor (HR)-positive, human epidermal growth factor receptor 2 (HER2)-negative breast cancer. In this review, we summarise the efficacy and toxicity findings from the randomised clinical trials, with simplified guidelines on the management of potential adverse effects. Education of healthcare professionals and patients is critical for safety and compliance. While there is some clinical evidence of activity of mTOR inhibition in HR-positive and HER2-positive breast cancers, the benefits may be more pronounced in selected subsets rather than in the overall population. Further development of predictive biomarkers will be useful in the selection of patients who will benefit from inhibition of the PI3K/Akt/mTOR (PAM) pathway.
Collapse
Affiliation(s)
- Joycelyn Jx Lee
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| | - Kiley Loh
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| | - Yoon-Sim Yap
- Department of Medical Oncology, National Cancer Center Singapore, Singapore 169610, Singapore
| |
Collapse
|
24
|
Timmermans-Sprang EPM, Gracanin A, Mol JA. High basal Wnt signaling is further induced by PI3K/mTor inhibition but sensitive to cSRC inhibition in mammary carcinoma cell lines with HER2/3 overexpression. BMC Cancer 2015. [PMID: 26205886 PMCID: PMC4513708 DOI: 10.1186/s12885-015-1544-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Elevated basal, ligand-independent, Wnt signaling in some canine breast cancer cells is not caused by classical mutations in APC, β-Catenin or GSK3β but, at least partially, by enhanced LEF1 expression. We examined the expression and function of EGFR/HER-regulated pathways on the ligand-independent Wnt signaling. Methods Twelve canine mammary tumor cell lines with previously reported differential basal Wnt activity were used. The expression levels of genes related to EGF-signaling were analyzed by cluster analysis. Cell lines with a combined overexpression of EGF-related genes and enhanced basal Wnt activity were treated with PI3K/mTor or cSRC inhibitors or transfected with a construct expressing wild-type PTEN. Subsequently, effects were measured on Wnt activity, cell proliferation, gene expression and protein level. Results High basal Wnt/LEF1 activity was associated with overexpression of HER2/3, ID1, ID2, RAC1 and HSP90 together with low to absent cMET and PTEN mRNA expression, suggesting a connection between Wnt- and HER-signaling pathways. Inhibition of the HER-regulated PI3K/mTor pathway using the dual PI3K/mTor inhibitor BEZ235 or the mTor inhibitor Everolimus® resulted in reduced cell proliferation. In the cell line with high basal Wnt activity, however, an unexpected further increased Wnt activity was found that could be greatly reduced after inhibition of the HER-regulated cSRC activity. Inhibition of the PI3K/mTor pathway was associated with enhanced expression of β-Catenin, Axin2, MUC1, cMET, EGFR and HER2 and a somewhat increased β-Catenin protein content, whereas cSRC inhibition was associated with slightly enhanced HER3 and SLUG mRNA expression. A high protein expression of HER3 was found only in a cell line with high basal Wnt activity. Conclusions High basal Wnt activity in some mammary cancer cell lines is associated with overexpression of HER-receptor related genes and HER3 protein, and the absence of PTEN. Inhibition of the PI3K/mTor pathway further stimulated, however, canonical Wnt signaling, whereas the inhibitory effect with the cSRC inhibitor Src-I1 on the Wnt activity further suggested a connection between Wnt and HER2/3-signaling.
Collapse
Affiliation(s)
- Elpetra P M Timmermans-Sprang
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584, CM, Utrecht, The Netherlands.
| | - Ana Gracanin
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584, CM, Utrecht, The Netherlands.
| | - Jan A Mol
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 104, 3584, CM, Utrecht, The Netherlands.
| |
Collapse
|
25
|
Sarma P, Bag I, Ramaiah MJ, Kamal A, Bhadra U, Pal Bhadra M. Bisindole-PBD regulates breast cancer cell proliferation via SIRT-p53 axis. Cancer Biol Ther 2015; 16:1486-501. [PMID: 26192233 DOI: 10.1080/15384047.2015.1071731] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In a previous study we reported the role of potent bisindole-PBD conjugate as an inclusion in the arsenal of breast cancer therapeutics. In breast cancer cell proliferation, PI3K/AKT/mTOR pathway plays a crucial role by prosurvival mechanism that inhibits programmed cell death. Here, 2 breast cancer cells lines, MCF-7 and MDA-MB-231 were treated with Vorinostat (suberoylanilide hydroxamic acid / SAHA) and bisindole-PBD (5b). We have investigated the effect on PI3K/AKT/mTOR pathway and SIRT expression including epigenetic regulation. There was consistent decrease in the level of PI3K, AKT, mTOR proteins upon treatment of 5b in both MCF-7 and MDA-MB-231 cell lines compared to untreated controls. Treatment with caspase inhibitor (Q-VD-OPH) confirmed that the effect of 5b on PI3K signaling was ahead of apoptosis. Real time PCR and western blot analysis showed profound reduction in the mRNA and protein levels of SIRT1 and SIRT2. Molecular docking studies also supported the interaction of 5b with various amino acids of SIRT2 proteins. Treatment with 5b caused epigenetic changes that include increase of acetylated forms of p53, increase of histone acetylation at p21 promoter as well as decrease in methylation state of p21 gene. Compound 5b thus acts as SIRT inhibitor and cause p53 activation via inhibition of growth factor signaling and activation of p53 dependent apoptotic signaling. This present study focuses bisindole-PBD on epigenetic alteration putting 5b as a promising therapeutic tool in the realm of breast cancer research.
Collapse
Affiliation(s)
- Pranjal Sarma
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| | - Indira Bag
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India.,b Functional Genomics and Gene Silencing Group; CSIR-Center for Cellular and Molecular Biology ; Hyderabad , India
| | - M Janaki Ramaiah
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India.,c School of Chemical & Biotechnology; SASTRA University ; Tirumalaisamudram, Thanjavur , India
| | - Ahmed Kamal
- d Medicinal Chemistry and Pharmacology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| | - Utpal Bhadra
- b Functional Genomics and Gene Silencing Group; CSIR-Center for Cellular and Molecular Biology ; Hyderabad , India
| | - Manika Pal Bhadra
- a Centre for Chemical Biology; CSIR-Indian Institute of Chemical Technology ; Tarnaka, Hyderabad , India
| |
Collapse
|
26
|
Wilks ST. Potential of overcoming resistance to HER2-targeted therapies through the PI3K/Akt/mTOR pathway. Breast 2015; 24:548-55. [PMID: 26187798 DOI: 10.1016/j.breast.2015.06.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 03/23/2015] [Accepted: 06/04/2015] [Indexed: 02/06/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression occurs in up to 30% of breast cancers and is a marker of aggressive disease. While HER2-targeted therapies have improved outcomes in these tumors, resistance to these agents develops in a large proportion of patients. Determining molecular mechanisms underlying resistance might help improve outcomes for patients with HER2-positive disease by allowing development of strategies to overcome resistance. Activation of signaling pathways involving the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway might contribute to the development of resistance to HER2-targeted therapies. Several inhibitors of this pathway are under investigation in this disease setting and phase 3 data for everolimus in combination with trastuzumab and chemotherapy in trastuzumab-refractory, advanced disease are promising. In this review, molecular mechanisms underlying resistance to HER2-targeted therapies are considered and evidence for strategies to manage resistance is evaluated, including the use of inhibitors of the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Sharon T Wilks
- Cancer Care Centers of South Texas, US Oncology, San Antonio, TX 78229, USA.
| |
Collapse
|
27
|
Inhibition of autophagy sensitizes malignant pleural mesothelioma cells to dual PI3K/mTOR inhibitors. Cell Death Dis 2015; 6:e1757. [PMID: 25950487 PMCID: PMC4669703 DOI: 10.1038/cddis.2015.124] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/26/2015] [Accepted: 03/30/2015] [Indexed: 01/01/2023]
Abstract
Malignant pleural mesothelioma (MPM) originates in most of the cases from chronic inflammation of the mesothelium due to exposure to asbestos fibers. Given the limited effect of chemotherapy, a big effort is being made to find new treatment options. The PI3K/mTOR pathway was reported to be upregulated in MPM. We tested the cell growth inhibition properties of two dual PI3K/mTOR inhibitors NVP-BEZ235 and GDC-0980 on 19 MPM cell lines. We could identify resistant and sensitive lines; however, there was no correlation to the downregulation of PI3K/mTOR activity markers. As a result of mTOR inhibition, both drugs efficiently induced long-term autophagy but not cell death. Autophagy blockade by chloroquine in combination with the dual PI3K/mTOR inhibitors significantly induced caspase-independent cell death involving RIP1 in the sensitive cell line SPC212. Cell death in the resistant cell line Mero-82 was less pronounced, and it was not induced via RIP1-dependent mechanism, suggesting the involvement of RIP1 downstream effectors. Cell death induction was confirmed in 3D systems. Based on these results, we identify autophagy as one of the main mechanisms of cell death resistance against dual PI3K/mTOR inhibitors in MPM. As PI3K/mTOR inhibitors are under investigation in clinical trials, these results may help interpreting their outcome and suggest ways for intervention.
Collapse
|
28
|
Sun Z, Li Q, Zhang S, Chen J, Huang L, Ren J, Chang Y, Liang Y, Wu G. NVP-BEZ235 overcomes gefitinib-acquired resistance by down-regulating PI3K/AKT/mTOR phosphorylation. Onco Targets Ther 2015; 8:269-77. [PMID: 25674002 PMCID: PMC4321659 DOI: 10.2147/ott.s62128] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Patients harboring activating mutations in epidermal growth factor receptors (EGFR) are particularly sensitive to EGFR tyrosine kinase inhibitors (TKIs). However, most patients develop an acquired resistance after a period of about 10 months. This study focuses on the therapeutic effect of NVP-BEZ235, a dual inhibitor of phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR), in gefitinib-resistant non-small cell lung cancer. Methods H1975 cell line was validated as a gefitinib-resistant cell model by the nucleotide-sequence analysis. We used the 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay to detect the growth of H1975 cell line in vitro. H1975 cells’ migration was detected by the migration assay. Xenograft models were used to investigate the growth of gefitinib-resistant non-small cell lung cancer in vivo. Western blot and immunohistochemical analysis were used to investigate the level of PI3K/protein kinase B(AKT)/mTOR signaling pathway proteins. Results We show that NVP-BEZ235 effectively inhibited the growth of H1975 cells in vivo as well as in vitro. Similarly, H1975 cell migration was reduced by NVP-BEZ235. Further experiments revealed that NVP-BEZ235 attenuated the phosphorylation of PI3K/AKT/mTOR signaling pathway proteins. Conclusion Taken together, we suggest that NVP-BEZ235 inhibits gefitinib-resistant tumor growth by downregulating PI3K/AKT/mTOR phosphorylation.
Collapse
Affiliation(s)
- Zhihua Sun
- Oncology department, Xiangyang central Hospital, Xiangyang, Hubei, People's Republic of China
| | - Qiuhui Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jing Chen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Lili Huang
- Radiation Oncology Department, Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, People's Republic of China
| | - Jinghua Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yu Chang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yichen Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| |
Collapse
|
29
|
Veigel D, Wagner R, Stübiger G, Wuczkowski M, Filipits M, Horvat R, Benhamú B, López-Rodríguez ML, Leisser A, Valent P, Grusch M, Hegardt FG, García J, Serra D, Auersperg N, Colomer R, Grunt TW. Fatty acid synthase is a metabolic marker of cell proliferation rather than malignancy in ovarian cancer and its precursor cells. Int J Cancer 2014; 136:2078-90. [DOI: 10.1002/ijc.29261] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 09/22/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Daniel Veigel
- Signaling Networks Program; Division of Oncology; Department of Medicine I; Medical University Vienna; Vienna Austria
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
| | - Renate Wagner
- Signaling Networks Program; Division of Oncology; Department of Medicine I; Medical University Vienna; Vienna Austria
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
| | - Gerald Stübiger
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
- Department of Vascular Biology and Thrombosis Research; Center of Physiology and Pharmacology, Medical University Vienna; Vienna Austria
| | - Michael Wuczkowski
- Department of Vascular Biology and Thrombosis Research; Center of Physiology and Pharmacology, Medical University Vienna; Vienna Austria
| | - Martin Filipits
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Vienna Austria
| | - Reinhard Horvat
- Clinical Institute of Pathology; Medical University of Vienna; Vienna Austria
| | - Bellinda Benhamú
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas, Universidad Complutense de Madrid; Madrid Spain
| | - María Luz López-Rodríguez
- Departamento de Química Orgánica I; Facultad de Ciencias Químicas, Universidad Complutense de Madrid; Madrid Spain
| | - Asha Leisser
- Signaling Networks Program; Division of Oncology; Department of Medicine I; Medical University Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Vienna Austria
| | - Peter Valent
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Vienna Austria
- Division of Hematology and Hemostaseology; Department of Medicine I; Medical University Vienna; Vienna Austria
| | - Michael Grusch
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
- Institute of Cancer Research; Department of Medicine I; Medical University of Vienna; Vienna Austria
| | - Fausto G. Hegardt
- Department of Biochemistry and Molecular Biology; Facultat de Farmàcia, Universitat de Barcelona; Barcelona Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB); Barcelona Spain
- CIBERobn Fisiopatología de la Obesidad y la Nutrición; Instituto de Salud Carlos III; Madrid Spain
| | - Jordi García
- Institut de Biomedicina de la Universitat de Barcelona (IBUB); Barcelona Spain
- CIBERobn Fisiopatología de la Obesidad y la Nutrición; Instituto de Salud Carlos III; Madrid Spain
- Department of Organic Chemistry; Facultat de Química; Universitat de Barcelona; Barcelona Spain
| | - Dolors Serra
- Department of Biochemistry and Molecular Biology; Facultat de Farmàcia, Universitat de Barcelona; Barcelona Spain
- Institut de Biomedicina de la Universitat de Barcelona (IBUB); Barcelona Spain
- CIBERobn Fisiopatología de la Obesidad y la Nutrición; Instituto de Salud Carlos III; Madrid Spain
| | - Nelly Auersperg
- Department of Obstetrics and Gynecology; University of British Columbia; Vancouver BC Canada
| | - Ramón Colomer
- Department of Medical Oncology; Hospital Universitario La Princesa; Madrid Spain
- Clinical Research Program; Spanish National Cancer Research Center (CNIO); Madrid Spain
| | - Thomas W. Grunt
- Signaling Networks Program; Division of Oncology; Department of Medicine I; Medical University Vienna; Vienna Austria
- Comprehensive Cancer Center, Medical University Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Vienna Austria
| |
Collapse
|
30
|
Fong P, Meng LR. Effect of mTOR inhibitors in nude mice with endometrial carcinoma and variable PTEN expression status. Med Sci Monit Basic Res 2014; 20:146-52. [PMID: 25266877 PMCID: PMC4189716 DOI: 10.12659/msmbr.892514] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background The aim of this study was to investigate the sensitivity to rapamycin of endometrial cancer cells with different phosphatase and tensin homologue (PTEN) expression to understand the mechanism of resistance to mammalian target of rapamycin (mTOR) inhibitors in the treatment of endometrial cancer. Material/Methods Twenty specific pathogen-free female BALB/c mice received transplants of either HEC-1A (PTEN-positive) or Ishikawa (PTEN-negative) cells. Mice in the treatment group were injected intraperitoneally once a week for 4 consecutive weeks. The control group was injected weekly with phosphate buffer saline (PBS) for 4 consecutive weeks. Tumor volume, tumor mass, growth curves, and inhibition rate were measured, after which the mice were killed. Results Both tumor growth rate and size were slower in the treatment group than in the control group for all mice that received transplants of either HEC-1A or Ishikawa cells. The tumor inhibition rates in the treatment group were 48.1% and 67.1% in mice transplanted with HEC-1A and Ishikawa cells, respectively. Conclusions The inhibitory effects of rapamycin were enhanced in PTEN-negative Ishikawa tumor cells compared with PTEN-positive HEC-1A cells, which could explain the reduced effect of rapalogues in some endometrial cancer patients and help to understand the mechanism of resistance to this drug.
Collapse
Affiliation(s)
- Pedro Fong
- School of Health Sciences, Macao Polytechnic Institute, Macao, China (mainland)
| | - Li-rong Meng
- School of Health Sciences, Macao Polytechnic Institute, Macao, China (mainland)
| |
Collapse
|
31
|
Gayle SS, Castellino RC, Buss MC, Nahta R. MEK inhibition increases lapatinib sensitivity via modulation of FOXM1. Curr Med Chem 2013; 20:2486-99. [PMID: 23531216 DOI: 10.2174/0929867311320190008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/07/2013] [Accepted: 03/12/2013] [Indexed: 12/22/2022]
Abstract
The standard targeted therapy for HER2-overexpressing breast cancer is the HER2 monoclonal antibody, trastuzumab. Although effective, many patients eventually develop trastuzumab resistance. The dual EGFR/HER2 small molecule tyrosine kinase inhibitor lapatinib is approved for use in trastuzumab-refractory metastatic HER2-positive breast cancer. However, lapatinib resistance is a problem as most patients with trastuzumab-refractory disease do not benefit from lapatinib. Understanding the mechanisms underlying lapatinib resistance may ultimately facilitate development of new therapeutic strategies for HER2-overexpressing breast cancer. Our current results indicate that MEK inhibition increases lapatinib-mediated cytotoxicity in resistant HER2-overexpressing breast cancer cells. We genetically and pharmacologically blocked MEK/ERK signaling and evaluated lapatinib response by trypan blue exclusion, anchorage-independent growth assays, flow cytometric cell cycle and apoptosis analysis, and in tumor xenografts. Combined MEK inhibition and lapatinib treatment reduced phosphorylated ERK more than single agent treatment. In addition, Western blots, immunofluorescence, and immunohistochemistry demonstrated that the combination of MEK inhibitor plus lapatinib reduced nuclear expression of the MEK/ERK downstream proto-oncogene FOXM1. Genetic knockdown of MEK was tested for the ability to increase lapatinib-mediated cell cycle arrest or apoptosis in JIMT-1 and MDA361 cells. Finally, xenograft studies demonstrated that combined pharmacological inhibition of MEK plus lapatinib suppressed tumor growth and reduced expression of FOXM1 in HER2-overexpressing breast cancers that are resistant to trastuzumab and lapatinib. Our results suggest that FoxM1 contributes to lapatinib resistance downstream of MEK signaling, and supports further study of pharmacological MEK inhibition to improve response to lapatinib in HER2-overexpressing trastuzumab-resistant breast cancer.
Collapse
Affiliation(s)
- S S Gayle
- Molecular & Systems Pharmacology Program, Graduate Division of Biological and Biomedical Sciences, Emory University, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
32
|
Abstract
The Rac inhibitor EHop-016 was developed as a compound with the potential to inhibit cancer metastasis. Inhibition of the first step of metastasis, migration, is an important strategy for metastasis prevention. The small GTPase Rac acts as a pivotal binary switch that is turned "on" by guanine nucleotide exchange factors (GEFs) via a myriad of cell surface receptors, to regulate cancer cell migration, survival, and proliferation. Unlike the related GTPase Ras, Racs are not usually mutated, but overexpressed or overactivated in cancer. Therefore, a rational Rac inhibitor should block the activation of Rac by its upstream effectors, GEFs, and the Rac inhibitor NSC23766 was developed using this rationale. However, this compound is ineffective at inhibiting the elevated Rac activity of metastatic breast cancer cells. Therefore, a panel of small molecule compounds were derived from NSC23766 and screened for Rac activity inhibition in metastatic cancer cells. EHop-016 was identified as a compound that blocks the interaction of Rac with the GEF Vav in metastatic human breast cancer cells with an IC50 of ~1μM. At higher concentrations (10μM), EHop-016 inhibits the related Rho GTPase Cdc42, but not Rho, and also reduces cell viability. Moreover, EHop-016 inhibits the activation of the Rac downstream effector p21-activated kinase, extension of motile actin-based structures, and cell migration. Future goals are to develop EHop-016 as a therapeutic to inhibit cancer metastasis, either individually or in combination with current anticancer compounds. The next generation of EHop-016-based Rac inhibitors is also being developed.
Collapse
Affiliation(s)
- Suranganie Dharmawardhane
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA.
| | - Eliud Hernandez
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA
| | - Cornelis Vlaar
- Department of Biochemistry, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico, USA
| |
Collapse
|
33
|
Martini M, Ciraolo E, Gulluni F, Hirsch E. Targeting PI3K in Cancer: Any Good News? Front Oncol 2013; 3:108. [PMID: 23658859 PMCID: PMC3647219 DOI: 10.3389/fonc.2013.00108] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/19/2013] [Indexed: 12/29/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) signaling pathway regulates several cellular processes and it’s one of the most frequently deregulated pathway in human tumors. Given its prominent role in cancer, there is great interest in the development of inhibitors able to target several members of PI3K signaling pathway in clinical trials. These drug candidates include PI3K inhibitors, both pan- and isoform-specific inhibitors, AKT, mTOR, and dual PI3K/mTOR inhibitors. As novel compounds progress into clinical trials, it’s becoming urgent to identify and select patient population that most likely benefit from PI3K inhibition. In this review we will discuss individual PIK3CA mutations as predictors of sensitivity and resistance to targeted therapies, leading to use of novel PI3K/mTOR/AKT inhibitors to a more “personalized” treatment.
Collapse
Affiliation(s)
- Miriam Martini
- Molecular Biotechnology Center, University of Turin Turin, Italy
| | | | | | | |
Collapse
|
34
|
Burris HA. Overcoming acquired resistance to anticancer therapy: focus on the PI3K/AKT/mTOR pathway. Cancer Chemother Pharmacol 2013; 71:829-42. [PMID: 23377372 DOI: 10.1007/s00280-012-2043-3] [Citation(s) in RCA: 342] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/23/2012] [Indexed: 12/23/2022]
Abstract
BACKGROUND Most targeted anticancer therapies, as well as cytotoxic and radiation therapies, are encumbered by the development of secondary resistance by cancer cells. Resistance is a complex phenomenon involving multiple mechanisms, including activation of signaling pathways such as phosphatidylinositol 3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR). Novel strategies to overcome resistance by targeting these signaling pathways are being evaluated. METHODS PubMed and key cancer congress abstracts were searched until July 2012 for preclinical and clinical data relating to the PI3K/AKT/mTOR pathway and anticancer treatment resistance, and use of PI3K/AKT/mTOR inhibitors in resistant cancer cell lines and patient populations. RESULTS Activation of the PI3K/AKT/mTOR pathway is frequently implicated in resistance to anticancer therapies, including biologics, tyrosine kinase inhibitors, radiation, and cytotoxics. As such, inhibitors of the PI3K/AKT/mTOR pathway are being rapidly evaluated in preclinical models and in clinical studies to determine whether they can restore therapeutic sensitivity when given in combination. In breast cancer, non-small-cell lung cancer, and glioblastoma, we find compelling preclinical evidence to show that inhibitors of PI3K or mTOR can restore sensitivity in resistant cells. Although clinical evidence is less mature, a recent Phase III study with the mTORC1 inhibitor everolimus in patients with advanced breast cancer resistant to aromatase inhibition and several Phase I/II studies with PI3K inhibitors demonstrate proof-of-concept, warranting future clinical evaluation. CONCLUSION Current preclinical and clinical evidence suggest that inhibitors of the PI3K/AKT/mTOR pathway could have utility in combination with other anticancer therapies to circumvent resistance by cancer cells. Multiple clinical studies are ongoing.
Collapse
Affiliation(s)
- Howard A Burris
- Sarah Cannon Research Institute, 3322 West End Avenue, Suite 900, Nashville, TN 37203, USA.
| |
Collapse
|
35
|
Cao Y, Chong Y, Shen H, Zhang M, Huang J, Zhu Y, Zhang Z. Combination of TNF-α and graphene oxide-loaded BEZ235 to enhance apoptosis of PIK3CA mutant colorectal cancer cells. J Mater Chem B 2013; 1:5602-5610. [DOI: 10.1039/c3tb20764a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
36
|
Korsse SE, Peppelenbosch MP, van Veelen W. Targeting LKB1 signaling in cancer. Biochim Biophys Acta Rev Cancer 2012; 1835:194-210. [PMID: 23287572 DOI: 10.1016/j.bbcan.2012.12.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 12/13/2022]
Abstract
The serine/threonine kinase LKB1 is a master kinase involved in cellular responses such as energy metabolism, cell polarity and cell growth. LKB1 regulates these crucial cellular responses mainly via AMPK/mTOR signaling. Germ-line mutations in LKB1 are associated with the predisposition of the Peutz-Jeghers syndrome in which patients develop gastrointestinal hamartomas and have an enormously increased risk for developing gastrointestinal, breast and gynecological cancers. In addition, somatic inactivation of LKB1 has been associated with sporadic cancers such as lung cancer. The exact mechanisms of LKB1-mediated tumor suppression remain so far unidentified; however, the inability to activate AMPK and the resulting mTOR hyperactivation has been detected in PJS-associated lesions. Therefore, targeting LKB1 in cancer is now mainly focusing on the activation of AMPK and inactivation of mTOR. Preclinical in vitro and in vivo studies show encouraging results regarding these approaches, which have even progressed to the initiation of a few clinical trials. In this review, we describe the functions, regulation and downstream signaling of LKB1, and its role in hereditary and sporadic cancers. In addition, we provide an overview of several AMPK activators, mTOR inhibitors and additional mechanisms to target LKB1 signaling, and describe the effect of these compounds on cancer cells. Overall, we will explain the current strategies attempting to find a way of treating LKB1-associated cancer.
Collapse
Affiliation(s)
- S E Korsse
- Dept. of Gastroenterology and Hepatology, Erasmus Medical University Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
37
|
Mohd Sharial MSN, Crown J, Hennessy BT. Overcoming resistance and restoring sensitivity to HER2-targeted therapies in breast cancer. Ann Oncol 2012; 23:3007-3016. [PMID: 22865781 PMCID: PMC3501233 DOI: 10.1093/annonc/mds200] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 05/03/2012] [Accepted: 05/14/2012] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Approximately 15%-23% of breast cancers overexpress human epidermal growth factor receptor 2 (HER2), which leads to the activation of signaling pathways that stimulate cell proliferation and survival. HER2-targeted therapy has substantially improved outcomes in patients with HER2-positive breast cancer. However, both de novo and acquired resistance are observed. DESIGN A literature search was performed to identify proposed mechanisms of resistance to HER2-targeted therapy and identified novel targets in clinical development for treating HER2-resistant disease. RESULTS Proposed HER2-resistance mechanisms include impediments to HER2-inhibitor binding, signaling through alternative pathways, upregulation of signaling pathways downstream of HER2, and failure to elicit an appropriate immune response. Although continuing HER2 inhibition beyond progression may provide an additional clinical benefit, the availability of novel therapies targeting different mechanisms of action could improve outcomes. The developmental strategy with the most available data is targeting the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (mTOR) pathway. The oral mTOR inhibitor everolimus has shown promising activity in combination with chemotherapy and trastuzumab in trastuzumab-refractory, advanced breast cancer. CONCLUSIONS Non-HER2-targeted therapy is a promising means of overcoming resistance to HER2-targeted treatment. Ongoing clinical studies will provide additional information on the efficacy and safety of novel targeted therapies in HER2-resistant advanced breast cancer.
Collapse
Affiliation(s)
- M S N Mohd Sharial
- Department of Medical Oncology, Beaumont Hospital, Dublin; Our Lady of Lourdes Hospital, Drogheda
| | - J Crown
- Department of Medical Oncology, St Vincent's University Hospital, Dublin, Ireland
| | - B T Hennessy
- Department of Medical Oncology, Beaumont Hospital, Dublin; Our Lady of Lourdes Hospital, Drogheda.
| |
Collapse
|
38
|
LoRusso PM. Mammalian target of rapamycin as a rational therapeutic target for breast cancer treatment. Oncology 2012; 84:43-56. [PMID: 23128843 DOI: 10.1159/000343063] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/27/2012] [Indexed: 12/16/2022]
Abstract
Therapies directed at endocrine receptors and human epidermal growth factor receptor 2 are important treatment options for patients with breast cancer; however, drug resistance and subsequent disease progression in patients with advanced disease is inevitable. The mammalian target of rapamycin (mTOR) is a key regulator of cell growth and proliferation implicated in the cellular processes that lead to the uncontrolled growth of cancer cells. Hence, overactivation of the mTOR pathway may also represent a key process in the development of resistance to these therapies, and interrupting this signaling cascade may alleviate resistance and help restore drug sensitivity. A number of agents that target the mTOR pathway have shown potent antitumorigenic effects in vitro, and several agents have also shown promise in treating patients with breast cancer. Everolimus and temsirolimus are the most clinically advanced agents in this class, with recent data from the BOLERO-2 study indicating significant benefit associated with everolimus when added to endocrine therapy in patients with endocrine therapy-resistant disease. In this review, we consider the translation of mTOR inhibitors from laboratory studies to large clinical trials, driven by a rational understanding of the role of mTOR in the processes that underlie breast cancer tumorigenesis.
Collapse
|
39
|
Current world literature. Curr Opin Oncol 2012; 24:756-68. [PMID: 23079785 DOI: 10.1097/cco.0b013e32835a4c91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
40
|
Cavazzoni A, Bonelli MA, Fumarola C, La Monica S, Airoud K, Bertoni R, Alfieri RR, Galetti M, Tramonti S, Galvani E, Harris AL, Martin LA, Andreis D, Bottini A, Generali D, Petronini PG. Overcoming acquired resistance to letrozole by targeting the PI3K/AKT/mTOR pathway in breast cancer cell clones. Cancer Lett 2012; 323:77-87. [DOI: 10.1016/j.canlet.2012.03.034] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/29/2012] [Accepted: 03/29/2012] [Indexed: 02/07/2023]
|
41
|
Fischer B, Frei C, Moura U, Stahel R, Felley-Bosco E. Inhibition of phosphoinositide-3 kinase pathway down regulates ABCG2 function and sensitizes malignant pleural mesothelioma to chemotherapy. Lung Cancer 2012; 78:23-9. [PMID: 22857894 DOI: 10.1016/j.lungcan.2012.07.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 06/21/2012] [Accepted: 07/08/2012] [Indexed: 12/12/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a relatively chemoresistant malignancy. Diverse biological targets are under investigation to develop new therapeutic options. One of these targets, namely the phosphoinositide-3-kinase (PI3K) pathway, has been shown to be a regulator of the side population (SP) phenotype in different cancers. The SP phenotype is due to drug efflux abilities providing drug-resistant properties. The presence of a SP fraction in MPM was recently observed in our laboratory. The aim of this study was to investigate the role of the PI3K pathway in the regulation of the SP phenotype in MPM. Treatment of overnight serum-starved cells with IGF increased phosphorylation of downstream target AKT, S6 and 4EBP1 and SP fraction in ZL55, ZL34 and SDM103T2 MPM cell lines. The PI3K/mTOR inhibitor NVP-BEZ235 and PI3K inhibitor wortmannin reduced the phosphorylation of downstream target AKT, S6 and 4EBP1 and decreased the SP fraction. Chemotherapy resistance mediated by drug efflux was tested by treating the cells with mitoxantrone. NVP-BEZ235 increased mitoxantrone cytotoxicity and this effect was mimicked by fumitremorgin C, a specific ABCG2 inhibitor, although not to the same extent, indicating that ABCG2-mediated drug efflux participates to chemoresistance. The involvement of ABCG2 in drug efflux was confirmed by determination of ABCG2-mediated decrease of intracellular mitoxantrone accumulation and silencing experiments. NVP-BEZ235-mediated decrease in drug efflux was associated with a significant decrease of ABCG2 present at the cell surface in ZL55 and SDM103T2 cells. In conclusion, the PI3K pathway is playing an important role in regulating the SP phenotype in MPM cells and inhibition of this activity may contribute to a more efficient cancer treatment.
Collapse
Affiliation(s)
- Bruno Fischer
- Molecular Oncology, Clinic for Oncology, University Hospital Zurich, Switzerland
| | | | | | | | | |
Collapse
|
42
|
Barbareschi M, Cuorvo LV, Girlando S, Bragantini E, Eccher C, Leonardi E, Ferro A, Caldara A, Triolo R, Cantaloni C, Decarli N, Galligioni E, Palma PD. PI3KCA mutations and/or PTEN loss in Her2-positive breast carcinomas treated with trastuzumab are not related to resistance to anti-Her2 therapy. Virchows Arch 2012; 461:129-39. [DOI: 10.1007/s00428-012-1267-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 05/28/2012] [Accepted: 06/13/2012] [Indexed: 12/19/2022]
|
43
|
Mueller A, Bachmann E, Linnig M, Khillimberger K, Schimanski CC, Galle PR, Moehler M. Selective PI3K inhibition by BKM120 and BEZ235 alone or in combination with chemotherapy in wild-type and mutated human gastrointestinal cancer cell lines. Cancer Chemother Pharmacol 2012; 69:1601-15. [PMID: 22543857 DOI: 10.1007/s00280-012-1869-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 04/12/2012] [Indexed: 01/09/2023]
Abstract
PURPOSE New targeted agents like antibodies or small molecules against tyrosine and lipid kinases clearly expand the standard therapy options in oncology. However, tumour resistance is still a challenge, often induced by mutations in growth-related signalling cascades. Twenty and ten percentage of all patients with colorectal and gastric cancers, respectively, carry phosphatidyl-3-kinase (PI3K) mutations and do not respond to receptor-blocking therapies. Recently, selective kinase inhibitors have been generated, which block the PI3K signalling pathway in tumour cells. So far, their therapeutic role for the treatment of mutated versus wild-type human gastrointestinal cancers has not been clarified in detail. METHODS To define the inhibitory and pro-apoptotic effects of the two PI3K inhibitors BEZ235 and BKM120 in three human colon cancer (HT-29, HCT-116 and DLD-1) and three gastric cancer (NCI-n87, AGS and MKN-45), cell lines with different PIK3CA gene mutation status were used. Firstly, viability, apoptosis and caspase assays were performed during incubation with either the inhibitors alone or combined with different cytotoxic agents. Secondly, the molecular consequences for the cell cycle and signalling pathways were analysed by defining the protein levels by FACS and Western blot analysis. RESULTS Both the PI3K inhibitors BEZ235 and BKM120 induced a clear concentration-dependent reduction in cell viability and an increase in apoptotic cell death, with the mutated cells being more sensitive to treatment. However, single-agent BEZ235 caused a G1 arrest in tumour cells, whilst BKM120 induced a G2 shift in a half of the gastrointestinal cancer cell lines. There was a clear downregulation in the protein levels of the PI3K-AKT pathway at the concentrations of 100 nM for both agents and for BEZ235 the additional inhibition of the mTOR pathway. Furthermore, BEZ235 caused synergistic induction of apoptosis when combined with irinotecan in colon cancer cell lines. Human gastric cancer cells were less sensitive to both BEZ235 and BKM120. CONCLUSIONS BEZ235 and BKM120 induced pro-apoptotic effects in all cell lines and especially with an increased response in the PI3KCA mutated cells. Our data support the clinical development of these PI3K inhibitors for patients with wild-type or mutated colon cancers.
Collapse
Affiliation(s)
- Annett Mueller
- First Department of Internal Medicine, University Medical Centre of the Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55101, Mainz, Germany.
| | | | | | | | | | | | | |
Collapse
|
44
|
Wilken JA, Badri T, Cross S, Raji R, Santin AD, Schwartz P, Branscum AJ, Baron AT, Sakhitab AI, Maihle NJ. EGFR/HER-targeted therapeutics in ovarian cancer. Future Med Chem 2012; 4:447-69. [PMID: 22416774 PMCID: PMC4620931 DOI: 10.4155/fmc.12.11] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Despite decades of research and evolving treatment modalities, survival among patients with epithelial ovarian cancer has improved only incrementally. During this same period, the development of biologically targeted therapeutics has improved survival for patients with diverse malignancies. Many of these new drugs target the human epidermal growth factor receptor (EGFR/HER/ErbB) family of tyrosine kinases, which play a major role in the etiology and progression of many carcinomas, including epithelial ovarian cancer. While several HER-targeted therapeutics are US FDA approved for the treatment of various malignancies, none have gained approval for the treatment of ovarian cancer. Here, we review the published literature on HER-targeted therapeutics for the treatment of ovarian cancer, including novel HER-targeted therapeutics in various stages of clinical development, as well as the challenges that have limited the use of these inhibitors in clinical settings.
Collapse
Affiliation(s)
- Jason A Wilken
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Tayf Badri
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Sarah Cross
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Rhoda Raji
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Alessandro D Santin
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Peter Schwartz
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Adam J Branscum
- Oregon State University, School of Biological & Population Health Sciences
| | - Andre T Baron
- University of Kentucky, Departments of Epidemiology, & Obstetrics & Gynecology
| | - Adam I Sakhitab
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
| | - Nita J Maihle
- Yale University, Department of Obstetrics, Gynecology & Reproductive Sciences
- Yale University, Departments of Pathology & Pharmacology
- PO Box 208063, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
45
|
Li J, Zhu F, Lubet RA, De Luca A, Grubbs C, Ericson ME, D'Alessio A, Normanno N, Dong Z, Bode AM. Quercetin-3-methyl ether inhibits lapatinib-sensitive and -resistant breast cancer cell growth by inducing G(2)/M arrest and apoptosis. Mol Carcinog 2011; 52:134-43. [PMID: 22086611 DOI: 10.1002/mc.21839] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/14/2011] [Accepted: 10/19/2011] [Indexed: 11/07/2022]
Abstract
Lapatinib, an oral, small-molecule, reversible inhibitor of both EGFR and HER2, is highly active in HER2 positive breast cancer as a single agent and in combination with other therapeutics. However, resistance against lapatinib is an unresolved problem in clinical oncology. Recently, interest in the use of natural compounds to prevent or treat cancers has gained increasing interest because of presumed low toxicity. Quercetin-3-methyl ether, a naturally occurring compound present in various plants, has potent anticancer activity. Here, we found that quercetin-3-methyl ether caused a significant growth inhibition of lapatinib-sensitive and -resistant breast cancer cells. Western blot data showed that quercetin-3-methyl ether had no effect on Akt or ERKs signaling in resistant cells. However, quercetin-3-methyl ether caused a pronounced G(2)/M block mainly through the Chk1-Cdc25c-cyclin B1/Cdk1 pathway in lapatinib-sensitive and -resistant cells. In contrast, lapatinib produced an accumulation of cells in the G(1) phase mediated through cyclin D1, but only in lapatinib-sensitive cells. Moreover, quercetin-3-methyl ether induced significant apoptosis, accompanied with increased levels of cleaved caspase 3, caspase 7, and poly(ADP-ribose) polymerase (PARP) in both cell lines. Overall, these results suggested that quercetin-3-methyl ether might be a novel and promising therapeutic agent in lapatinib-sensitive or -resistant breast cancer patients.
Collapse
Affiliation(s)
- Jixia Li
- The Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Tomek K, Wagner R, Varga F, Singer CF, Karlic H, Grunt TW. Blockade of Fatty Acid Synthase Induces Ubiquitination and Degradation of Phosphoinositide-3-Kinase Signaling Proteins in Ovarian Cancer. Mol Cancer Res 2011; 9:1767-79. [DOI: 10.1158/1541-7786.mcr-10-0467] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
47
|
Roper J, Richardson MP, Wang WV, Richard LG, Chen W, Coffee EM, Sinnamon MJ, Lee L, Chen PC, Bronson RT, Martin ES, Hung KE. The dual PI3K/mTOR inhibitor NVP-BEZ235 induces tumor regression in a genetically engineered mouse model of PIK3CA wild-type colorectal cancer. PLoS One 2011; 6:e25132. [PMID: 21966435 PMCID: PMC3180374 DOI: 10.1371/journal.pone.0025132] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Accepted: 08/25/2011] [Indexed: 01/08/2023] Open
Abstract
Purpose To examine the in vitro and in vivo efficacy of the dual PI3K/mTOR inhibitor NVP-BEZ235 in treatment of PIK3CA wild-type colorectal cancer (CRC). Experimental Design PIK3CA mutant and wild-type human CRC cell lines were treated in vitro with NVP-BEZ235, and the resulting effects on proliferation, apoptosis, and signaling were assessed. Colonic tumors from a genetically engineered mouse (GEM) model for sporadic wild-type PIK3CA CRC were treated in vivo with NVP-BEZ235. The resulting effects on macroscopic tumor growth/regression, proliferation, apoptosis, angiogenesis, and signaling were examined. Results In vitro treatment of CRC cell lines with NVP-BEZ235 resulted in transient PI3K blockade, sustained decreases in mTORC1/mTORC2 signaling, and a corresponding decrease in cell viability (median IC50 = 9.0–14.3 nM). Similar effects were seen in paired isogenic CRC cell lines that differed only in the presence or absence of an activating PIK3CA mutant allele. In vivo treatment of colonic tumor-bearing mice with NVP-BEZ235 resulted in transient PI3K inhibition and sustained blockade of mTORC1/mTORC2 signaling. Longitudinal tumor surveillance by optical colonoscopy demonstrated a 97% increase in tumor size in control mice (p = 0.01) vs. a 43% decrease (p = 0.008) in treated mice. Ex vivo analysis of the NVP-BEZ235-treated tumors demonstrated a 56% decrease in proliferation (p = 0.003), no effects on apoptosis, and a 75% reduction in angiogenesis (p = 0.013). Conclusions These studies provide the preclinical rationale for studies examining the efficacy of the dual PI3K/mTOR inhibitor NVP-BEZ235 in treatment of PIK3CA wild-type CRC.
Collapse
Affiliation(s)
- Jatin Roper
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Michael P. Richardson
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Wei Vivian Wang
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Larissa Georgeon Richard
- Department of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wei Chen
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Erin M. Coffee
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Mark J. Sinnamon
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Lydia Lee
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
| | - Peng-Chieh Chen
- Department of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Roderick T. Bronson
- Dana Farber/Harvard Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eric S. Martin
- Dana Farber/Harvard Cancer Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kenneth E. Hung
- Division of Gastroenterology, Department of Medicine, Tufts Medical Center, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
48
|
PI3K pathway activation results in low efficacy of both trastuzumab and lapatinib. BMC Cancer 2011; 11:248. [PMID: 21676217 PMCID: PMC3141770 DOI: 10.1186/1471-2407-11-248] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Accepted: 06/15/2011] [Indexed: 12/11/2022] Open
Abstract
Background Human epidermal growth factor receptor 2 (HER2) is the most crucial ErbB receptor tyrosine kinase (RTK) family member in HER2-positive (refered to HER2-overexpressing) breast cancer which are dependent on or "addictive" to the Phosphatidylinositol-3-kinase (PI3K) pathway. HER2-related target drugs trastuzumab and lapatinib have been the foundation of treatment of HER2--positive breast cancer. This study was designed to explore the relationship between PI3K pathway activation and the sensitivity to lapatinib in HER2--positive metastatic breast cancer patients pretreated with anthracyclins, taxanes and trastuzumab. Methods Sixty-seven HER2-positive metastatic breast cancer patients were recruited into a global lapatinib Expanded Access Program and 57 patients have primary tumor specimens available for determination of PI3K pathway status. PTEN status was determined by immunohistochemical staining and PIK3CA mutations were detected via PCR sequencing. All patients were treated with lapatinib 1250 mg/day continuously and capecitabine 1000 mg/m2 twice daily on a 2-week-on and 1-week-off schedule until disease progression, death, withdrawal of informed consent, or intolerable toxicity. Results PIK3CA mutations and PTEN loss were detected in 12.3% (7/57) and 31.6% (18/57) of the patients, respectively. Twenty-two patients with PI3K pathway activation (defined as PIK3CA mutation and/or PTEN expression loss) had a lower clinical benefit rate (36.4% versus 68.6%, P = 0.017) and a lower overall response rate (9.1% versus 31.4%, P = 0.05), when compared with the 35 patients with no activation. A retrospective analysis of first trastuzumab-containing regimen treatment data showed that PI3K pathway activation correlated with a shorter median progression-free survival (4.5 versus 9.0 months, P = 0.013). Conclusions PIK3CA mutations occur more frequently in elder patients for HER2-positive breast cancer. PIK3CA mutations and PTEN loss are not mutually exclusive. PI3K pathway activation resulting from PTEN loss or PIK3CA mutations may lead to drug resistance to lapatinib and trastuzumab (http://ClinicalTrials.gov number, NCT00338247).
Collapse
|
49
|
Munagala R, Aqil F, Gupta RC. Promising molecular targeted therapies in breast cancer. Indian J Pharmacol 2011; 43:236-45. [PMID: 21713084 PMCID: PMC3113372 DOI: 10.4103/0253-7613.81497] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 02/23/2011] [Indexed: 12/27/2022] Open
Abstract
In recent years, there has been a significant improvement in the understanding of molecular events and critical pathways involved in breast cancer. This has led to the identification of novel targets and development of anticancer therapies referred to as targeted therapy. Targeted therapy has high specificity for the molecules involved in key molecular events that are responsible for cancer phenotype such as cell growth, survival, migration, invasion, metastasis, apoptosis, cell-cycle progression, and angiogenesis. Targeted agents that have been approved for breast cancer include trastuzumab and lapatinib, directed against human epidermal growth factor receptor 2 (HER2) and bevacizumab, directed against vascular endothelial growth factor (VEGF). Several other targeted agents currently under evaluation in preclinical and clinical trials include inhibitors of epidermal growth factor receptor (EGFR), dual EGFR and HER2 inhibitors, VEGF/VEGFR inhibitors, and agents that interfere with crucial signaling pathways such as PI3K/AKT/mTOR and RAS/MEK/ERK; agents against other tyrosine kinases such as Src, insulin-like growth factor (IGF)/IGF-receptor (IGFR); agents that promote apoptosis such as Poly ADP ribose polymerase inhibitors; agents that target invasion and metastasis such as matrix metalloproteinases inhibitors and others. In this review, we highlight the most promising targeted agents and their combination with mainstream chemotherapeutic drugs in clinical trials.
Collapse
Affiliation(s)
- Radha Munagala
- James Graham Brown Cancer Center, University of Louisville, Louisville
| | - Farrukh Aqil
- James Graham Brown Cancer Center, University of Louisville, Louisville
| | - Ramesh C. Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville
- Department of Pharmacology and Toxicology, University of Louisville, Louisville
| |
Collapse
|