1
|
Kwon YS, Nam KS, Kim S. Tamoxifen overcomes the trastuzumab-resistance of SK-BR-3 tumorspheres by targeting crosstalk between cytoplasmic estrogen receptor α and the EGFR/HER2 signaling pathway. Biochem Pharmacol 2021; 190:114635. [PMID: 34058187 DOI: 10.1016/j.bcp.2021.114635] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/17/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Since trastuzumab-resistance remains a major obstacle to the successful treatment of HER2-positive breast cancer, a detailed understanding of the mechanisms responsible is required to direct future pharmacotherapeutic strategies. Recently, several studies have indicated that the quiescent natures of cancer stem cells contribute to treatment resistance and tumor recurrence. Thus, in this study, we investigated the mechanism underlying trastuzumab resistance in a quiescent cell population using tumorsphere cultures and explored better therapeutic strategies to overcome trastuzumab resistance in HER2-positive breast cancer patients. We observed that most cells in SK-BR-3 tumorspheres were quiescent, showing the accumulation of cells at the G0/G1 phase as compared to cells in monolayer culture. Furthermore, SK-BR-3 tumorspheres exhibited enhanced EGFR/HER2 signaling, which was incompletely inhibited by trastuzumab, and subsequently led to trastuzumab-resistance. Interestingly, cytoplasmic estrogen receptor α (ERα) expression was markedly elevated in tumorspheres and was associated with enhanced EGFR/HER2 signaling. Accordingly, inhibition of ERα with tamoxifen selectively targeted tumorspheres rather than cells in monolayer culture and overcame trastuzumab resistance in tumorspheres. Taken together, our findings indicate that crosstalk between cytoplasmic ERα and the HER2/EGFR signaling pathway can be considered a novel therapeutic target for quiescent cell populations within HER2-positive breast cancer and that simultaneous inhibition of ER and the EGFR/HER2 pathway may prevent trastuzumab resistance. We hope that these results provide a basis for the use of combinations of tamoxifen and trastuzumab in HER2-positive breast cancer patients.
Collapse
Affiliation(s)
- Yun-Suk Kwon
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Dongdae-ro 123, Gyeongju, Gyeongsangbuk-do 38066, Republic of Korea
| | - Kyung-Soo Nam
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Dongdae-ro 123, Gyeongju, Gyeongsangbuk-do 38066, Republic of Korea
| | - Soyoung Kim
- Department of Pharmacology and Intractable Disease Research Center, School of Medicine, Dongguk University, Dongdae-ro 123, Gyeongju, Gyeongsangbuk-do 38066, Republic of Korea.
| |
Collapse
|
2
|
Abedin MR, Powers K, Aiardo R, Barua D, Barua S. Antibody-drug nanoparticle induces synergistic treatment efficacies in HER2 positive breast cancer cells. Sci Rep 2021; 11:7347. [PMID: 33795712 PMCID: PMC8016985 DOI: 10.1038/s41598-021-86762-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
Chemotherapeutic drugs suffer from non-specific binding, undesired toxicity, and poor blood circulation which contribute to poor therapeutic efficacy. In this study, antibody–drug nanoparticles (ADNs) are engineered by synthesizing pure anti-cancer drug nanorods (NRs) in the core of nanoparticles with a therapeutic monoclonal antibody, Trastuzumab on the surface of NRs for specific targeting and synergistic treatments of human epidermal growth factor receptor 2 (HER2) positive breast cancer cells. ADNs were designed by first synthesizing ~ 95 nm diameter × ~ 500 nm long paclitaxel (PTX) NRs using the nanoprecipitation method. The surface of PTXNRs was functionalized at 2′ OH nucleophilic site using carbonyldiimidazole and conjugated to TTZ through the lysine residue interaction forming PTXNR-TTZ conjugates (ADNs). The size, shape, and surface charge of ADNs were characterized using scanning electron microscopy (SEM), SEM, and zeta potential, respectively. Using fluorophore labeling and response surface analysis, the percentage conjugation efficiency was found > 95% with a PTX to TTZ mass ratio of 4 (molar ratio ≈ 682). In vitro therapeutic efficiency of PTXNR-TTZ was evaluated in two HER2 positive breast cancer cell lines: BT-474 and SK-BR-3, and a HER2 negative MDA-MB-231 breast cancer cell using MTT assay. PTXNR-TTZ inhibited > 80% of BT-474 and SK-BR-3 cells at a higher efficiency than individual PTX and TTZ treatments alone after 72 h. A combination index analysis indicated a synergistic combination of PTXNR-TTZ compared with the doses of single-drug treatment. Relatively lower cytotoxicity was observed in MCF-10A human breast epithelial cell control. The molecular mechanisms of PTXNR-TTZ were investigated using cell cycle and Western blot analyses. The cell cycle analysis showed PTXNR-TTZ arrested > 80% of BT-474 breast cancer cells in the G2/M phase, while > 70% of untreated cells were found in the G0/G1 phase indicating that G2/M arrest induced apoptosis. A similar percentage of G2/M arrested cells was found to induce caspase-dependent apoptosis in PTXNR-TTZ treated BT-474 cells as revealed using Western blot analysis. PTXNR-TTZ treated BT-474 cells showed ~ 1.3, 1.4, and 1.6-fold higher expressions of cleaved caspase-9, cytochrome C, and cleaved caspase-3, respectively than untreated cells, indicating up-regulation of caspase-dependent activation of apoptotic pathways. The PTXNR-TTZ ADN represents a novel nanoparticle design that holds promise for targeted and efficient anti-cancer therapy by selective targeting and cancer cell death via apoptosis and mitotic cell cycle arrest.
Collapse
Affiliation(s)
- Muhammad Raisul Abedin
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Kaitlyne Powers
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Rachel Aiardo
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA
| | - Dibbya Barua
- Department of Electrical and Computer Engineering, Missouri University of Science and Technology, Rolla, MO, 65409, USA
| | - Sutapa Barua
- Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, 110 Bertelsmeyer Hall, 1101 N. State Street, Rolla, MO, 65409-1230, USA.
| |
Collapse
|
3
|
Combined Targeting of Estrogen Receptor Alpha and XPO1 Prevent Akt Activation, Remodel Metabolic Pathways and Induce Autophagy to Overcome Tamoxifen Resistance. Cancers (Basel) 2019; 11:cancers11040479. [PMID: 30987380 PMCID: PMC6520695 DOI: 10.3390/cancers11040479] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/29/2019] [Accepted: 03/29/2019] [Indexed: 01/21/2023] Open
Abstract
A majority of breast cancer specific deaths in women with ERα (+) tumors occur due to metastases that are resistant to endocrine therapy. There is a critical need for novel therapeutic approaches to resensitize recurrent ERα (+) tumors to endocrine therapies. The objective of this study was to elucidate mechanisms of improved effectiveness of combined targeting of ERα and the nuclear transport protein XPO1 in overcoming endocrine resistance. Selinexor (SEL), an XPO1 antagonist, has been evaluated in multiple late stage clinical trials in patients with relapsed and /or refractory hematological and solid tumor malignancies. Our transcriptomics analysis showed that 4-Hydroxytamoxifen (4-OHT), SEL alone or their combination induced differential Akt signaling- and metabolism-associated gene expression profiles. Western blot analysis in endocrine resistant cell lines and xenograft models validated differential Akt phosphorylation. Using the Seahorse metabolic profiler, we showed that ERα-XPO1 targeting changed the metabolic phenotype of TAM-resistant breast cancer cells from an energetic to a quiescent profile. This finding demonstrated that combined targeting of XPO1 and ERα rewired the metabolic pathways and shut down both glycolytic and mitochondrial pathways that would eventually lead to autophagy. Remodeling metabolic pathways to regenerate new vulnerabilities in endocrine resistant breast tumors is novel, and given the need for better strategies to improve therapy response in relapsed ERα (+) tumors, our findings show great promise for uncovering the role that ERα-XPO1 crosstalk plays in reducing cancer recurrences.
Collapse
|
4
|
Trastuzumab decorated TPGS-g-chitosan nanoparticles for targeted breast cancer therapy. Colloids Surf B Biointerfaces 2019; 173:366-377. [DOI: 10.1016/j.colsurfb.2018.10.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/27/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023]
|
5
|
Wang M, Hu Y, Yu T, Ma X, Wei X, Wei Y. Pan-HER-targeted approach for cancer therapy: Mechanisms, recent advances and clinical prospect. Cancer Lett 2018; 439:113-130. [PMID: 30218688 DOI: 10.1016/j.canlet.2018.07.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/08/2018] [Accepted: 07/09/2018] [Indexed: 02/05/2023]
Abstract
The Human Epidermal Growth Factor Receptor family is composed of 4 structurally related receptor tyrosine kinases that are involved in many human cancers. The efficacy and safety of HER inhibitors have been compared in a wide range of clinical trials, suggesting the superior inhibitory ability of multiple- HER-targeting blockade compared with single receptor antagonists. However, many patients are currently resistant to current therapeutic treatment and novel strategies are warranted to conquer the resistance. Thus, we performed a critical review to summarize the molecular involvement of HER family receptors in tumour progression, recent anti-HER drug development based on clinical trials, and the potential resistance mechanisms of anti-HER therapy.
Collapse
Affiliation(s)
- Manni Wang
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Yuzhu Hu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Ting Yu
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xuelei Ma
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| | - Xiawei Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China.
| | - Yuquan Wei
- Lab of Aging Research and Nanotoxicology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, PR China
| |
Collapse
|
6
|
Lee JH, Son YH, Kwon YW, Park SY, Koo BS, Jung SH. Anticancer Effects of a Korean Herbal Medicine Formula (H9) via AMPK and HER2-PI3K/Akt Signaling in Breast Cancer Cells. Phytother Res 2017; 31:1765-1775. [DOI: 10.1002/ptr.5906] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 07/18/2017] [Accepted: 08/07/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Ju-Hee Lee
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Young Hoon Son
- Department of Biochemistry, College of Medicine; Seoul National University; Seoul 03080 Korea
| | - Young-Won Kwon
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Sung Yun Park
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Byung-Soo Koo
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| | - Seung Hyun Jung
- College of Korean Medicine; Dongguk University; Gyeongju 38066 Korea
| |
Collapse
|
7
|
Chakraborty A, Hatzis C, DiGiovanna MP. Co-targeting the HER and IGF/insulin receptor axis in breast cancer, with triple targeting with endocrine therapy for hormone-sensitive disease. Breast Cancer Res Treat 2017; 163:37-50. [PMID: 28236033 DOI: 10.1007/s10549-017-4169-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 02/17/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE Interactions between HER2, estrogen receptor (ER), and insulin-like growth factor I receptor (IGF1R) are implicated in resistance to monotherapies targeting these receptors. We have previously shown in pre-clinical studies synergistic anti-tumor effects for co-targeting each pairwise combination of HER2, IGF1R, and ER. Strikingly, synergy for HER2/IGF1R targeting occurred not only in a HER2+ model, but also in a HER2-normal model. The purpose of the current study was therefore to determine the generalizability of synergistic anti-tumor effects of co-targeting HER2/IGF1R, the anti-tumor activity of triple-targeting HER2/IGF1R/ER in hormone-dependent cell lines, and the effect of using the multi-targeting drugs neratinib (pan-HER) and BMS-754807 (dual IGF1R/insulin receptor). METHODS Proliferation and apoptosis assays were performed in a large panel of cell lines representing varying receptor expression levels. Mechanistic effects were studied using phospho-protein immunoblotting. Analyses of drug interaction effects were performed using linear mixed-effects regression models. RESULTS Enhanced anti-proliferative effects of HER/IGF-insulin co-targeting were seen in most, though not all, cell lines, including HER2-normal lines. For ER+ lines, triple targeting with inclusion of anti-estrogen generally resulted in the greatest anti-tumor effects. Double or triple targeting generally resulted in marked increases in apoptosis in the sensitive lines. Mechanistic studies demonstrated that the synergy between drugs was correlated with maximal inhibition of Akt and ERK pathway signaling. CONCLUSIONS Dual HER/IGF-insulin targeting, and triple targeting with inclusion of anti-estrogen drugs, shows striking anti-tumor activity across breast cancer types, and drugs with broader receptor specificity may be more effective than single receptor selective drugs, particularly for ER- cells.
Collapse
Affiliation(s)
- Ashok Chakraborty
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Christos Hatzis
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | - Michael P DiGiovanna
- Section of Medical Oncology, Departments of Internal Medicine, Yale Cancer Center, Smilow Cancer Hospital, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA.
| |
Collapse
|
8
|
Richards JO, Albers AJ, Smith TS, Tjoe JA. NK cell-mediated antibody-dependent cellular cytotoxicity is enhanced by tamoxifen in HER2/neu non-amplified, but not HER2/neu-amplified, breast cancer cells. Cancer Immunol Immunother 2016; 65:1325-1335. [PMID: 27573917 PMCID: PMC11028446 DOI: 10.1007/s00262-016-1885-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 08/08/2016] [Indexed: 01/06/2023]
Abstract
Tumor-targeting antibodies have been successful in the treatment of various types of cancers. Antibodies engage the immune system with their Fc, stimulating immune cell effector function. In the clinic, FcγRIIIa polymorphisms with higher affinity for the Fc of antibodies were shown to improve response rates and overall survival. Efforts have been made to modify the Fc to enhance affinity to Fc receptors and thereby improve effector function. An alternative for improving immune effector function may be to increase the level of tumor antigen expression. In this study, tamoxifen was used to increase HER2/neu protein level to determine whether increased tumor antigen expression could enhance NK cell-mediated antibody-dependent cytotoxicity (ADCC). Tamoxifen was found to increase HER2/neu 1.5-fold to threefold in breast cancer cell lines that were HER2/neu non-amplified. Using flow cytometry to simultaneously evaluate NK cell degranulation and tumor cell death, the increase in HER2/neu enhanced NK cell-mediated ADCC. However, in cells that had HER2/neu gene amplification and estrogen receptor expression, tamoxifen elevated HER2/neu but failed to improve NK cell function. The quantity of HER2/neu on the tumor cell surface was approximately double that of the number of Fc receptors found on NK cells. This appears to reflect a ceiling at which increasing antigen expression fails to improve NK cell effector function. This has clinical implications as trying to increase antigen expression to enhance NK cell function may be useful for patients with antigen-low tumors, but not in those whose tumors have gene amplification or high levels of antigen expression.
Collapse
Affiliation(s)
- John O Richards
- Immunotherapy Research Laboratory, Aurora St. Luke's Medical Center, Aurora Health Care, Milwaukee, WI, USA.
- Aurora Research Institute, Aurora Health Care, Milwaukee, WI, USA.
- Immunology Research Laboratory, Aurora Research Institute, 960 N. 12th Street, 3rd Floor, Milwaukee, WI, 53233, USA.
| | - Alex J Albers
- Immunotherapy Research Laboratory, Aurora St. Luke's Medical Center, Aurora Health Care, Milwaukee, WI, USA
- Aurora Research Institute, Aurora Health Care, Milwaukee, WI, USA
| | - Thomas S Smith
- Immunotherapy Research Laboratory, Aurora St. Luke's Medical Center, Aurora Health Care, Milwaukee, WI, USA
- Aurora Research Institute, Aurora Health Care, Milwaukee, WI, USA
| | - Judy A Tjoe
- Aurora Research Institute, Aurora Health Care, Milwaukee, WI, USA
- Surgical Breast Oncology, Aurora Cancer Care, Aurora Health Care, Milwaukee, WI, USA
| |
Collapse
|
9
|
Wrobel K, Zhao YC, Kulkoyluoglu E, Chen KLA, Hieronymi K, Holloway J, Li S, Ray T, Ray PS, Landesman Y, Lipka AE, Smith RL, Madak-Erdogan Z. ERα-XPO1 Cross Talk Controls Tamoxifen Sensitivity in Tumors by Altering ERK5 Cellular Localization. Mol Endocrinol 2016; 30:1029-1045. [PMID: 27533791 PMCID: PMC5045498 DOI: 10.1210/me.2016-1101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/12/2016] [Indexed: 12/22/2022] Open
Abstract
Most breast cancer deaths occur in women with recurrent, estrogen receptor (ER)-α(+), metastatic tumors. There is a critical need for therapeutic approaches that include novel, targetable mechanism-based strategies by which ERα (+) tumors can be resensitized to endocrine therapies. The objective of this study was to validate a group of nuclear transport genes as potential biomarkers to predict the risk of endocrine therapy failure and to evaluate the inhibition of XPO1, one of these genes as a novel means to enhance the effectiveness of endocrine therapies. Using advanced statistical methods, we found that expression levels of several of nuclear transport genes including XPO1 were associated with poor survival and predicted recurrence of tamoxifen-treated breast tumors in human breast cancer gene expression data sets. In mechanistic studies we showed that the expression of XPO1 determined the cellular localization of the key signaling proteins and the response to tamoxifen. We demonstrated that combined targeting of XPO1 and ERα in several tamoxifen-resistant cell lines and tumor xenografts with the XPO1 inhibitor, Selinexor, and tamoxifen restored tamoxifen sensitivity and prevented recurrence in vivo. The nuclear transport pathways have not previously been implicated in the development of endocrine resistance, and given the need for better strategies for selecting patients to receive endocrine modulatory reagents and improving therapy response of relapsed ERα(+) tumors, our findings show great promise for uncovering the role these pathways play in reducing cancer recurrences.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Hormonal/pharmacology
- Biological Transport/drug effects
- Biological Transport/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cell Nucleus/drug effects
- Cell Nucleus/genetics
- Cell Nucleus/metabolism
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Estrogen Receptor alpha/genetics
- Estrogen Receptor alpha/metabolism
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Karyopherins/genetics
- Karyopherins/metabolism
- MCF-7 Cells
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Mitogen-Activated Protein Kinase 7/genetics
- Mitogen-Activated Protein Kinase 7/metabolism
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Tamoxifen/pharmacology
- Exportin 1 Protein
Collapse
Affiliation(s)
- Kinga Wrobel
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yiru Chen Zhao
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Eylem Kulkoyluoglu
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Karen Lee Ann Chen
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Kadriye Hieronymi
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Jamie Holloway
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Sarah Li
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Tania Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Partha Sarathi Ray
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Yosef Landesman
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Alexander Edward Lipka
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Rebecca Lee Smith
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| | - Zeynep Madak-Erdogan
- Department of Food Science and Human Nutrition (K.W., Y.C.Z., E.K., K.H., Z.M.-E.), Division of Nutritional Sciences (K.L.A.C., Z.M.-E.), University of Illinois at Urbana-Champaign, Departments of Surgery (P.S.R.) and Bioengineering (P.S.R.), Interdisciplinary Health Sciences Institute (P.S.R.), and Division of Surgical Oncology (P.S.R.), Carle Cancer Center, and Departments of Crop Sciences (A.E.L.) and Pathobiology (R.L.S.), College of Veterinary Medicine, Urbana, Illinois 61801; (J.H.), Arlington, Massachusetts; Onconostic Technologies Inc (S.L., T.R., P.S.R.), Urbana, Illinois 61820; Karyopharm Therapeutics (Y.L.), Newton, Massachusetts 02459; and Cancer Community Illinois (Z.M.-E.), Urbana, Illinois 61801
| |
Collapse
|
10
|
Yin L, Wang ZY. Roles of the ER-α36-EGFR/HER2 positive regulatory loops in tamoxifen resistance. Steroids 2016; 111:95-99. [PMID: 26884313 DOI: 10.1016/j.steroids.2016.01.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/28/2016] [Accepted: 01/28/2016] [Indexed: 11/16/2022]
Abstract
Tamoxifen provided a successful treatment for ER-positive breast cancer for the past four decades. However, most breast tumors are eventually resistant to tamoxifen therapy. Extensive researches were conducted to understand the molecular mechanisms involved in tamoxifen resistance, and have revealed that multiple signaling molecules and pathways such as EGFR and HER2 are involved in tamoxifen resistance. Currently, the mechanisms by which tamoxifen sensitive breast cancer cells acquire these signaling pathways and develop tamoxifen resistance have not been elucidated. The identification of ER-α36, a variant of ER-α, that is able to mediate agonist activity of tamoxifen provided great insights into the underlying mechanisms of tamoxifen resistance. In this review, we will discuss the biological function and the possible underlying mechanisms of ER-α36 in tamoxifen resistance and specifically illustrate a novel cross-talk mechanism; positive regulatory loops between the ER-α36 and EGFR/HER2 in tamoxifen resistance. The function and the underlying mechanisms of ER-α36 in tamoxifen resistance of the breast cancer stem/progenitor cells will also be discussed. Finally, we will postulate a novel approach to restore tamoxifen sensitivity in tamoxifen resistant breast cancer cells.
Collapse
Affiliation(s)
- Li Yin
- Department of Medical Microbiology and Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE, USA
| | - Zhao-Yi Wang
- Department of Medical Microbiology and Immunology, Creighton University Medical School, 2500 California Plaza, Omaha, NE, USA.
| |
Collapse
|
11
|
Sini V, Cinieri S, Conte P, De Laurentiis M, Leo AD, Tondini C, Marchetti P. Endocrine therapy in post-menopausal women with metastatic breast cancer: From literature and guidelines to clinical practice. Crit Rev Oncol Hematol 2016; 100:57-68. [PMID: 26944782 DOI: 10.1016/j.critrevonc.2016.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/15/2016] [Indexed: 12/22/2022] Open
Abstract
Current international guidelines recommend endocrine therapy as the initial treatment of choice in hormone receptor positive advanced breast cancer. Endocrine therapy has been a mainstay of hormone responsive breast cancer treatment for more than a century. To date it is based on different approaches,such as blocking the estrogen receptor through selective receptor estrogen modulators, depleting extragonadal peripheral estrogen synthesis by aromatase inhibitors or inducing estrogen receptor degradation using selective down-regulators. Despite estrogen and/or progesterone receptor positive status, up to a quarter of patients could be either primarily resistant to hormone therapies or will develop hormone resistance during the course of their disease. Different mechanisms, either intrinsic or acquired, could be implicated in endocrine resistance. In the present work available endocrine therapies and their appropriate sequences have been reviewed, and the most promising strategies to overcome endocrine resistance have been highlighted.
Collapse
Affiliation(s)
- Valentina Sini
- Surgical and Medical Department of Clinical Sciences, Biomedical Technologies and Translational Medicine, "Sapienza" University of Rome, Italy; Oncology Department, Santo Spirito Hospital, Rome, Italy
| | - Saverio Cinieri
- Medical Oncology Department & Breast Unit-Hospital of Brindisi and Medical Oncology Department-European Institute of Oncology, Milan, Italy
| | - Pierfranco Conte
- Medical Oncology 2, Venetian Oncological Institute, Padova, Italy; Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | - Michelino De Laurentiis
- Department of Breast Oncology, National Cancer Institute "Fondazione Pascale", Naples, Italy
| | - Angelo Di Leo
- Medical Oncology Department, "Sandro Pitigliani" Hospital of Prato, Istituto Toscano Tumori, Prato, Italy
| | - Carlo Tondini
- USC Oncologia Medica, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Marchetti
- Department of Clinical and Molecular Medicine, Medical Oncology Division, Sant'Andrea Hospital, "Sapienza" University of Rome, Rome, Italy; IDI-IRCCS, Rome, Italy.
| |
Collapse
|
12
|
Abstract
The majority of metastatic breast cancers cannot be cured and present a major public health problem worldwide. Approximately 70% of breast cancers express the estrogen receptor, and endocrine-based therapies have significantly improved patient outcomes. However, the development of endocrine resistance is extremely common. Understanding the molecular pathways that regulate the hormone sensitivity of breast cancer cells is important to improving the efficacy of endocrine therapy. It is becoming clearer that the PI3K-AKT-forkhead box O (FOXO) signaling axis is a key player in the hormone-independent growth of many breast cancers. Constitutive PI3K-AKT pathway activation, a driver of breast cancer growth, causes down-regulation of FOXO tumor suppressor functions. This review will summarize what is currently known about the role of FOXOs in endocrine-resistance mechanisms. It will also suggest potential therapeutic strategies for the restoration of normal FOXO transcriptional activity.
Collapse
Affiliation(s)
- M Bullock
- Hormones and Cancer GroupCancer Genetics Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, Pacific Highway Saint Leonards, Sydney, New South Wales 2065, Australia
| |
Collapse
|
13
|
Tamayo P, Steinhardt G, Liberzon A, Mesirov JP. The limitations of simple gene set enrichment analysis assuming gene independence. Stat Methods Med Res 2016; 25:472-87. [PMID: 23070592 PMCID: PMC3758419 DOI: 10.1177/0962280212460441] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Since its first publication in 2003, the Gene Set Enrichment Analysis method, based on the Kolmogorov-Smirnov statistic, has been heavily used, modified, and also questioned. Recently a simplified approach using a one-sample t-test score to assess enrichment and ignoring gene-gene correlations was proposed by Irizarry et al. 2009 as a serious contender. The argument criticizes Gene Set Enrichment Analysis's nonparametric nature and its use of an empirical null distribution as unnecessary and hard to compute. We refute these claims by careful consideration of the assumptions of the simplified method and its results, including a comparison with Gene Set Enrichment Analysis's on a large benchmark set of 50 datasets. Our results provide strong empirical evidence that gene-gene correlations cannot be ignored due to the significant variance inflation they produced on the enrichment scores and should be taken into account when estimating gene set enrichment significance. In addition, we discuss the challenges that the complex correlation structure and multi-modality of gene sets pose more generally for gene set enrichment methods.
Collapse
Affiliation(s)
- Pablo Tamayo
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - George Steinhardt
- Boston University Bioinformatics Program, Boston University, Boston, MA, USA
| | - Arthur Liberzon
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Jill P Mesirov
- The Eli and Edythe L. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA Boston University Bioinformatics Program, Boston University, Boston, MA, USA
| |
Collapse
|
14
|
Mason JK, Klaire S, Kharotia S, Wiggins AKA, Thompson LU. α-linolenic acid and docosahexaenoic acid, alone and combined with trastuzumab, reduce HER2-overexpressing breast cancer cell growth but differentially regulate HER2 signaling pathways. Lipids Health Dis 2015; 14:91. [PMID: 26282560 PMCID: PMC4539855 DOI: 10.1186/s12944-015-0090-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 07/31/2015] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Diets rich in the n-3 fatty acid alpha-linolenic acid (ALA) have been shown to reduce breast tumor growth, enhance the effectiveness of the HER2-targeted drug trastuzumab (TRAS) and reduce HER2 signaling in mouse models. It is unclear whether this is due to direct effects of ALA or due to its long-chain n-3 fatty acids metabolites including docosahexaenoic acid (DHA). METHODS The ability of HER2-overexpressing BT-474 human breast cancer cells to convert ALA to long-chain n-3 fatty acids was determined by measurement of phospholipid fatty acids by gas chromatography following treatment with 100 μM ALA. The effects of 96 h treatment with ALA or DHA, at serum levels seen in mice (50-100 μM), alone and combined with TRAS (10 μg/ml), on BT-474 cell growth measured by trypan blue exclusion, apoptosis measured by flow cytometric analysis of Annexin-V/7-AAD stained cells (ALA and TRAS treatment only) and protein biomarkers HER2 signaling measured by western blot were determined. RESULTS ALA-treated BT-474 cells had higher phospholipid ALA but no increase in downstream n-3 metabolites including DHA. Both ALA and DHA reduced cell growth with and without TRAS. ALA had no effect on apoptosis. ALA and DHA showed opposite effects on Akt and MAPK phosphorylation; ALA increased and DHA decreased phosphorylation. CONCLUSIONS Together these data suggest that, while both ALA and its DHA metabolite can reduce HER2-overexpressing breast cancer growth with and without TRAS, they demonstrate for the first time that DHA is responsible for the effects of ALA-rich diets on HER2 signaling pathways.
Collapse
Affiliation(s)
- Julie K Mason
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College Street, Toronto, ON, M5S 3E2, Canada.
| | - Sukhpreet Klaire
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College Street, Toronto, ON, M5S 3E2, Canada.
| | - Shikhil Kharotia
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College Street, Toronto, ON, M5S 3E2, Canada.
| | - Ashleigh K A Wiggins
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College Street, Toronto, ON, M5S 3E2, Canada.
| | - Lilian U Thompson
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, 150 College Street, Toronto, ON, M5S 3E2, Canada.
| |
Collapse
|
15
|
Chakraborty AK, Zerillo C, DiGiovanna MP. In vitro and in vivo studies of the combination of IGF1R inhibitor figitumumab (CP-751,871) with HER2 inhibitors trastuzumab and neratinib. Breast Cancer Res Treat 2015. [PMID: 26195122 DOI: 10.1007/s10549-015-3504-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The insulin-like growth factor I receptor (IGF1R) has been linked to resistance to HER2-directed therapy with trastuzumab (Herceptin). We examined the anti-tumor activity of figitumumab (CP-751,871), a human monoclonal antibody that blocks IGF1R ligand binding, alone and in combination with the therapeutic anti-HER2 antibody trastuzumab and the pan-HER family tyrosine kinase inhibitor neratinib, using in vitro and in vivo breast cancer model systems. In vitro assays of proliferation, apoptosis, and signaling, and in vivo anti-tumor experiments were conducted in HER2-overexpressing (BT474) and HER2-normal (MCF7) models. We find single-agent activity of the HER2-targeting drugs but not figitumumab in the BT474 model, while the reverse is true in the MCF7 model. However, in both models, combining figitumumab with HER2-targeting drugs shows synergistic anti-proliferative and apoptosis-inducing effects, and optimum inhibition of downstream signaling. In murine xenograft models, synergistic anti-tumor effects were observed in the HER2-normal MCF7 model for the combination of figitumumab with trastuzumab, and, in the HER2-overexpressing BT474 model, enhanced anti-tumor effects were observed for the combination of figitumumab with either trastuzumab or neratinib. Analysis of tumor extracts from the in vivo experiments showed evidence of the most optimal inhibition of downstream signaling for the drug combinations over the single-agent therapies. These results suggest promise for such combinations in treating patients with breast cancer, and that, unlike the case for single-agent therapy, the therapeutic effects of such combinations may be independent of expression levels of the individual receptors or the single-agent activity profile.
Collapse
Affiliation(s)
- Ashok K Chakraborty
- Departments of Internal Medicine (Section of Medical Oncology) and Pharmacology, and The Yale Cancer Center, Yale University School of Medicine, 300 George Street, Suite 120, New Haven, CT, 06510, USA
| | | | | |
Collapse
|
16
|
Zhou Y, Gou LT, Guo ZH, Liu HR, Wang JM, Zhou SX, Yang JL, Li XA. Fully human HER2/cluster of differentiation 3 bispecific antibody triggers potent and specific cytotoxicity of T lymphocytes against breast cancer. Mol Med Rep 2015; 12:147-54. [PMID: 25760691 PMCID: PMC4438947 DOI: 10.3892/mmr.2015.3441] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 02/10/2015] [Indexed: 02/05/2023] Open
Abstract
The use of a bispecific antibody (BsAb) is a promising and highly specific approach to cancer therapy. In the present study, a fully human recombinant single chain variable fragment BsAb against human epidermal growth factor receptor (HER)2 and cluster of differentiation (CD)3 was constructed with the aim of developing an effective treatment for breast cancer. HER2/CD3 BsAb was expressed in Chinese hamster ovary cells and purified via nickel column chromatography. Flow cytometry revealed that the HER2/CD3 BsAb was able to specifically bind to HER2 and CD3-positive cells. HER2/CD3 BsAb was able to stimulate T-cell activation and induce the lysis of cultured SKBR-3 and BT474 cells in the presence of unstimulated T lymphocytes. HER2/CD3 BsAb efficiently inhibited the growth of breast cancer tissue by activating and inducing the proliferation of tumor tissue infiltrating lymphocytes. Therefore, HER2/CD3 BsAb is a potent tool which may be a suitable candidate for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yan Zhou
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Lan-Tu Gou
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zhi-Hui Guo
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Hai-Rong Liu
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Jiang-Man Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Shu-Xian Zhou
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| | - Jin-Liang Yang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiao-An Li
- Gastroenterology Tumor and Microenvironment Laboratory, Department of Gastroenterology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
17
|
Off-target-free gene delivery by affinity-purified receptor-targeted viral vectors. Nat Commun 2015; 6:6246. [PMID: 25665714 DOI: 10.1038/ncomms7246] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 01/08/2015] [Indexed: 11/09/2022] Open
Abstract
We describe receptor-targeted adeno-associated viral (AAV) vectors that allow genetic modification of rare cell types ex vivo and in vivo while showing no detectable off-targeting. Displaying designed ankyrin repeat proteins (DARPins) on the viral capsid and carefully depleting DARPin-deficient particles, AAV vectors were made specific for Her2/neu, EpCAM or CD4. A single intravenous administration of vector targeted to the tumour antigen Her2/neu was sufficient to track 75% of all tumour sites and to extend survival longer than the cytostatic antibody Herceptin. CD4-targeted AAVs hit human CD4-positive cells present in spleen of a humanized mouse model, while CD8-positive cells as well as liver or other off-target organs remained unmodified. Mimicking conditions of circulating tumour cells, EpCAM-AAV detected single tumour cells in human blood opening the avenue for tumour stem cell tracking. Thus, the approach developed here delivers genes to target cell types of choice with antibody-like specificity.
Collapse
|
18
|
Mason JK, Fu M, Chen J, Thompson LU. Flaxseed oil enhances the effectiveness of trastuzumab in reducing the growth of HER2-overexpressing human breast tumors (BT-474). J Nutr Biochem 2015; 26:16-23. [DOI: 10.1016/j.jnutbio.2014.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/17/2014] [Accepted: 08/01/2014] [Indexed: 11/25/2022]
|
19
|
Regina A, Demeule M, Tripathy S, Lord-Dufour S, Currie JC, Iddir M, Annabi B, Castaigne JP, Lachowicz JE. ANG4043, a Novel Brain-Penetrant Peptide–mAb Conjugate, Is Efficacious against HER2-Positive Intracranial Tumors in Mice. Mol Cancer Ther 2014; 14:129-40. [PMID: 25492620 DOI: 10.1158/1535-7163.mct-14-0399] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
LIU MEI, YANG YAJUN, ZHENG HONG, ZHONG XIAORONG, WANG YU, WANG ZHU, WANG YAOGENG, WANG YANPING. Membrane-bound complement regulatory proteins are prognostic factors of operable breast cancer treated with adjuvant trastuzumab: A retrospective study. Oncol Rep 2014; 32:2619-27. [DOI: 10.3892/or.2014.3496] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/20/2014] [Indexed: 11/06/2022] Open
|
21
|
Nagy Z, Kovács I, Török M, Tóth D, Vereb G, Buzás K, Juhász I, Blumberg PM, Bíró T, Czifra G. Function of RasGRP3 in the formation and progression of human breast cancer. Mol Cancer 2014; 13:96. [PMID: 24779681 PMCID: PMC4113147 DOI: 10.1186/1476-4598-13-96] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/21/2014] [Indexed: 02/07/2023] Open
Abstract
Introduction Ras guanine nucleotide exchange factors (RasGEFs) mediate the activation of the Ras signaling pathway that is over activated in many human cancers. The RasGRP3, an activator of H-Ras and R-Ras protein exerts oncogenic effects and the overexpression of the protein is observed in numerous malignant cancer types. Here, we investigated the putative alteration of expression and potential function of RasGRP3 in the formation and progression of human breast cancer. Methods The RasGRP3 and phosphoRasGRP3 expressions were examined in human invasive ductal adenocarcinoma derived samples and cell lines (BT-474, JIMT-1, MCF7, SK-BR-3, MDA-MB-453, T-47D) both in mRNA (Q-PCR) and protein (Western blot; immunohistochemistry) levels. To explore the biological function of the protein, RasGRP3 knockdown cultures were established. To assess the role of RasGRP3 in the viability of cells, annexin-V/PI staining and MitoProbe™ DilC1 (5) assay were performed. To clarify the function of the protein in cell proliferation and in the development of chemotherapeutic resistance, CyQuant assay was performed. To observe the RasGRP3 function in tumor formation, the Severe combined immunodeficiency (SCID) mouse model was used. To investigate the role of the protein in Ras-related signaling Q-PCR and Western blot experiments were performed. Results RasGRP3 expression was elevated in human breast tumor tissue samples as well as in multiple human breast cancer cell lines. Down-regulation of RasGRP3 expression in breast cancer cells decreased cell proliferation, induced apoptosis in MCF7 cells, and sensitized T-47D cells to the action of drugs Tamoxifen and trastuzumab (Herceptin). Gene silencing of RasGRP3 reduced tumor formation in mouse xenografts as well. Inhibition of RasGRP3 expression also reduced Akt, ERK1/2 and estrogen receptor alpha phosphorylation downstream from IGF-I insulin like growth factor-I (IGF-I) or epidermal growth factor (EGF) stimulation confirming the functional role of RasGRP3 in the altered behavior of these cells. Conclusions Taken together, our results suggest that the Ras activator RasGRP3 may have a role in the pathological behavior of breast cancer cells and may constitute a therapeutic target for human breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Gabriella Czifra
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, University of Debrecen, Medical and Health Science Center, Research Center for Molecular Medicine, Nagyerdei krt, 98, PO Box 22, Debrecen H-4032, Hungary.
| |
Collapse
|
22
|
Barrett JS, Gupta M, Mondick JT. Model-based drug development applied to oncology. Expert Opin Drug Discov 2013; 2:185-209. [PMID: 23496077 DOI: 10.1517/17460441.2.2.185] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Model-based drug development (MBDD) is an approach that is used to organize the vast and complex data streams that feed the drug development pipelines of small molecule and biotechnology sponsors. Such data streams are ultimately reviewed by the global regulatory community as evidence of a drug's potential to treat and/or harm patients. Some of this information is captured in the scientific literature and prescribing compendiums forming the basis of how new and existing agents will ultimately be administered and further evaluated in the broader patient community. As this data stream evolves, the details of data qualification, the assumptions and/or critical decisions based on these data are lost under conventional drug development paradigms. MBDD relies on the construction of quantitative relationships to connect data from discrete experiments conducted along the drug development pathway. These relationships are then used to ask questions relevant at critical development stages, hopefully, with the understanding that the various scenarios explored represent a path to optimal decision making. Oncology, as a therapeutic area, presents a unique set of challenges and perhaps a different development paradigm as opposed to other disease targets. The poor attrition of development compounds in the recent past attests to these difficulties and provides an incentive for a different approach. In addition, given the reliance on multimodal therapy, oncological disease targets are often treated with both new and older agents spanning several drug classes. As MBDD becomes more integrated into the pharmaceutical research community, a more rational explanation for decisions regarding the development of new oncology agents as well as the proposed treatment regimens that incorporate both new and existing agents can be expected. Hopefully, the end result is a more focussed clinical development programme, which ultimately provides a means to optimize individual patient care.
Collapse
Affiliation(s)
- Jeffrey S Barrett
- Laboratory for Applied PK/PD, Clinical Pharmacology & Therapeutics Division, The Children's Hospital of Philadelphia, USA .
| | | | | |
Collapse
|
23
|
Ceran C, Cokol M, Cingoz S, Tasan I, Ozturk M, Yagci T. Novel anti-HER2 monoclonal antibodies: synergy and antagonism with tumor necrosis factor-α. BMC Cancer 2012; 12:450. [PMID: 23033967 PMCID: PMC3517359 DOI: 10.1186/1471-2407-12-450] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 09/11/2012] [Indexed: 11/13/2022] Open
Abstract
Background One-third of breast cancers display amplifications of the ERBB2 gene encoding the HER2 kinase receptor. Trastuzumab, a humanized antibody directed against an epitope on subdomain IV of the extracellular domain of HER2 is used for therapy of HER2-overexpressing mammary tumors. However, many tumors are either natively resistant or acquire resistance against Trastuzumab. Antibodies directed to different epitopes on the extracellular domain of HER2 are promising candidates for replacement or combinatorial therapy. For example, Pertuzumab that binds to subdomain II of HER2 extracellular domain and inhibits receptor dimerization is under clinical trial. Alternative antibodies directed to novel HER2 epitopes may serve as additional tools for breast cancer therapy. Our aim was to generate novel anti-HER2 monoclonal antibodies inhibiting the growth of breast cancer cells, either alone or in combination with tumor necrosis factor-α (TNF-α). Methods Mice were immunized against SK-BR-3 cells and recombinant HER2 extracellular domain protein to produce monoclonal antibodies. Anti-HER2 antibodies were characterized with breast cancer cell lines using immunofluorescence, flow cytometry, immunoprecipitation, western blot techniques. Antibody epitopes were localized using plasmids encoding recombinant HER2 protein variants. Antibodies, either alone or in combination with TNF-α, were tested for their effects on breast cancer cell proliferation. Results We produced five new anti-HER2 monoclonal antibodies, all directed against conformational epitope or epitopes restricted to the native form of the extracellular domain. When tested alone, some antibodies inhibited modestly but significantly the growth of SK-BR-3, BT-474 and MDA-MB-361 cells displaying ERBB2 amplification. They had no detectable effect on MCF-7 and T47D cells lacking ERBB2 amplification. When tested in combination with TNF-α, antibodies acted synergistically on SK-BR-3 cells, but antagonistically on BT-474 cells. A representative anti-HER2 antibody inhibited Akt and ERK1/2 phosphorylation leading to cyclin D1 accumulation and growth arrest in SK-BR-3 cells, independently from TNF-α. Conclusions Novel antibodies against extracellular domain of HER2 may serve as potent anti-cancer bioactive molecules. Cell-dependent synergy and antagonism between anti-HER2 antibodies and TNF-α provide evidence for a complex interplay between HER2 and TNF-α signaling pathways. Such complexity may drastically affect the outcome of HER2-directed therapeutic interventions.
Collapse
Affiliation(s)
- Ceyhan Ceran
- BilGen Genetics and Biotechnology Research Center, Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | | | | | | | | | | |
Collapse
|
24
|
Huang HC, Lin CL, Lin JK. 1,2,3,4,6-penta-O-galloyl-β-D-glucose, quercetin, curcumin and lycopene induce cell-cycle arrest in MDA-MB-231 and BT474 cells through downregulation of Skp2 protein. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:6765-6775. [PMID: 21598989 DOI: 10.1021/jf201096v] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
The F-box protein S-phase kinase-associated protein 2 (Skp2), which acts as an oncogene through targeting p27 for degradation, is overexpressed in many different human cancers. Skp2 can play an important role in breast cancer progression and may also be a novel molecular target for the treatment of breast cancer, especially estrogen receptor (ER)/human epidermal growth factor 2 (HER2) negative breast cancers. Unfortunately, specific drugs that target Skp2 are unavailable at present. Therefore, it is important to explore whether commonly used chemopreventive agents may downregulate Skp2 expression. In this study, we examined the effects of 1,2,3,4,6-penta-O-galloyl-β-D-glucose (pentagalloylglucose, 5gg), quercetin, curcumin and lycopene on the expression of Skp2 in MDA-MB-231 (ER/HER2-negative) and BT474 (ER-negative/HER2-positive) cells. We found that all four phytochemicals studied induced cell growth inhibition in MDA-MB-231 cells. The mechanism of the initial growth inhibitory events involves blocking the cell cycle progression. Further, we found that quercetin and curcumin induced growth arrest by inhibition of Skp2, and induced p27 expression in MDA-MB-231 cells. However, the decrease in Skp2 levels in cells treated with 5gg or lycopene did not translate to p27 upregulation. Consequently, the downregulation of Skp2 did not always correlate with the upregulation of p27, suggesting that phytochemical-dependent downregulation of Skp2 can influence cell growth in several ways. Several studies have demonstrated that Skp2 directs the ubiquitylation and subsequent degradation of forkhead box protein O1 (FoxO1). Furthermore, our results reveal that FoxO1 protein was increased after 5gg, quercetin, curcumin and lycopene treatment. The therapeutic strategies designed to reduce Skp2 may therefore play an important clinical role in treatment of breast cancer cells, especially ER/HER2-negative breast cancers.
Collapse
Affiliation(s)
- Hsiu-Chen Huang
- Department of Applied Science, National Hsinchu University of Education, Hsinchu 30014, Taiwan
| | | | | |
Collapse
|
25
|
Liang K, Esteva FJ, Albarracin C, Stemke-Hale K, Lu Y, Bianchini G, Yang CY, Li Y, Li X, Chen CT, Mills GB, Hortobagyi GN, Mendelsohn J, Hung MC, Fan Z. Recombinant human erythropoietin antagonizes trastuzumab treatment of breast cancer cells via Jak2-mediated Src activation and PTEN inactivation. Cancer Cell 2010; 18:423-35. [PMID: 21075308 PMCID: PMC3022383 DOI: 10.1016/j.ccr.2010.10.025] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Revised: 07/23/2010] [Accepted: 09/10/2010] [Indexed: 12/24/2022]
Abstract
We found that the receptor for erythropoietin (EpoR) is coexpressed with human epidermal growth factor receptor-2 (HER2) in a significant percentage of human breast tumor specimens and breast cancer cell lines. Exposure of HER2 and EpoR dual-positive breast cancer cells to recombinant human erythropoietin (rHuEPO) activated cell signaling. Concurrent treatment of the cells with rHuEPO and trastuzumab reduced the cells' response to trastuzumab both in vitro and in vivo. We identified Jak2-mediated activation of Src and inactivation of PTEN as underlying mechanisms through which rHuEPO antagonizes trastuzumab-induced therapeutic effects. Furthermore, we found that compared with administration of trastuzumab alone, concurrent administration of rHuEPO and trastuzumab correlated with shorter progression-free and overall survival in patients with HER2-positive metastatic breast cancer.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Drug Antagonism
- Drug Resistance, Neoplasm/drug effects
- Enzyme Activation
- Erythropoietin/pharmacology
- Erythropoietin/therapeutic use
- Female
- Humans
- Janus Kinase 2/physiology
- Mice
- PTEN Phosphohydrolase/metabolism
- Proto-Oncogene Proteins pp60(c-src)/metabolism
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptors, Erythropoietin/chemistry
- Receptors, Erythropoietin/metabolism
- Recombinant Proteins
- Signal Transduction
- Transplantation, Heterologous
- Trastuzumab
Collapse
Affiliation(s)
- Ke Liang
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Francisco J. Esteva
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Constance Albarracin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Katherine Stemke-Hale
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yang Lu
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Giampaolo Bianchini
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ching-Yi Yang
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yong Li
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xinqun Li
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chun-Te Chen
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gordon B. Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John Mendelsohn
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Center for Molecular Medicine and Graduate Institute of Cancer Biology, China Medical University and Hospital, Taichung, Taiwan
| | - Zhen Fan
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Correspondence:
| |
Collapse
|
26
|
Koay DC, Zerillo C, Narayan M, Harris LN, DiGiovanna MP. Anti-tumor effects of retinoids combined with trastuzumab or tamoxifen in breast cancer cells: induction of apoptosis by retinoid/trastuzumab combinations. Breast Cancer Res 2010; 12:R62. [PMID: 20696059 PMCID: PMC2949655 DOI: 10.1186/bcr2625] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Revised: 05/21/2010] [Accepted: 08/09/2010] [Indexed: 12/31/2022] Open
Abstract
INTRODUCTION HER2 and estrogen receptor (ER) are important in breast cancer and are therapeutic targets of trastuzumab (Herceptin) and tamoxifen, respectively. Retinoids inhibit breast cancer growth, and modulate signaling by HER2 and ER. We hypothesized that treatment with retinoids and simultaneous targeting of HER2 and/or ER may have enhanced anti-tumor effects. METHODS The effects of retinoids combined with trastuzumab or tamoxifen were examined in two human breast cancer cell lines in culture, BT474 and SKBR3. Assays of proliferation, apoptosis, differentiation, cell cycle distribution, and receptor signaling were performed. RESULTS In HER2-overexpressing/ER-positive BT474 cells, combining all-trans retinoic acid (atRA) with tamoxifen or trastuzumab synergistically inhibited cell growth, and altered cell differentiation and cell cycle. Only atRA/trastuzumab-containing combinations induced apoptosis. BT474 and HER2-overexpressing/ER-negative SKBR3 cells were treated with a panel of retinoids (atRA, 9-cis-retinoic acid, 13-cis-retinoic acid, or N-(4-hydroxyphenyl) retinamide (fenretinide) (4-HPR)) combined with trastuzumab. In BT474 cells, none of the single agents except 4-HPR induced apoptosis, but again combinations of each retinoid with trastuzumab did induce apoptosis. In contrast, the single retinoid agents did cause apoptosis in SKBR3 cells; this was only modestly enhanced by addition of trastuzumab. The retinoid drug combinations altered signaling by HER2 and ER. Retinoids were inactive in trastuzumab-resistant BT474 cells. CONCLUSIONS Combining retinoids with trastuzumab maximally inhibits cell growth and induces apoptosis in trastuzumab-sensitive cells. Treatment with such combinations may have benefit for breast cancer patients.
Collapse
Affiliation(s)
- Debbie C Koay
- Department of Internal Medicine (Section of Medical Oncology), Yale Cancer Center and Smilow Cancer Hospital at Yale-New-Haven Hospital, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
27
|
Sabnis G, Brodie A. Understanding Resistance to Endocrine Agents: Molecular Mechanisms and Potential for Intervention. Clin Breast Cancer 2010; 10:E6-E15. [DOI: 10.3816/cbc.2010.n.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
28
|
Ryu WS, Son GS. Cancer Vaccines Targeting HER2/neu for Early Breast Cancer. J Breast Cancer 2010. [DOI: 10.4048/jbc.2010.13.1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Woo Sang Ryu
- Department of Surgery, Ansan Hospital, Korea University School of Medicine, Ansan, Korea
| | - Gil Soo Son
- Department of Surgery, Ansan Hospital, Korea University School of Medicine, Ansan, Korea
| |
Collapse
|
29
|
Chakraborty AK, Welsh A, Digiovanna MP. Co-targeting the insulin-like growth factor I receptor enhances growth-inhibitory and pro-apoptotic effects of anti-estrogens in human breast cancer cell lines. Breast Cancer Res Treat 2009; 120:327-35. [PMID: 19337828 DOI: 10.1007/s10549-009-0382-5] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 03/16/2009] [Indexed: 11/25/2022]
Abstract
The insulin-like growth factor I receptor (IGF1R) interacts with estrogen receptor-alpha (ERalpha) and HER2. We examined the effect of combinations of IGF1R antagonists (alpha-IR3, AG1024) and anti-estrogens (4-hydroxy tamoxifen, fulvestrant) in two human ER+ breast cancer cell lines: BT474 (HER2 overexpressing, IGF1R low) and MCF7 (HER2 non-overexpressing, IGF1R high). In BT474 cells, growth was inhibited by anti-estrogens, but not by IGF1R antagonists; however, adding IGF1R inhibitors to anti-estrogens enhanced growth inhibition. In MCF7 cells, growth was inhibited by IGF1R and ER antagonists and more so by their combination. In both cell lines, no single agents could induce apoptosis, but combining IGF1R inhibitors with anti-estrogens induced dramatic levels of apoptosis. IGF1R antagonists enhanced the ability of the anti-estrogens to inhibit ER transcriptional activity in BT474 cells, but not in MCF7 cells. The drug combination synergistically inhibited ER and IGF1R activity. Such combinations may be useful therapy for breast cancer.
Collapse
Affiliation(s)
- Ashok K Chakraborty
- Departments of Internal Medicine (Section of Medical Oncology) and Pharmacology, and the Yale Cancer Center, Yale University School of Medicine, 333 Cedar Street, Room WWW217, New Haven, CT 06510, USA
| | | | | |
Collapse
|
30
|
The ErbB inhibitors trastuzumab and erlotinib inhibit growth of vestibular schwannoma xenografts in nude mice: a preliminary study. Otol Neurotol 2009; 29:846-53. [PMID: 18636037 DOI: 10.1097/mao.0b013e31817f7398] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To analyze the ability of ErbB inhibitors to reduce the growth of vestibular schwannoma (VS) xenografts. METHODS Vestibular schwannoma xenografts were established in the interscapular fat pad in nude mice for 4 weeks. Initially, a small cohort of animals was treated with the ErbB2 inhibitor trastuzumab or saline for 2 weeks. Animals also received bromodeoxyuridine injections to label proliferating cells. In a longer-term experiment, animals were randomized to receive trastuzumab, erlotinib (an ErbB kinase inhibitor), or placebo for 12 weeks. Tumor growth was monitored by magnetic resonance imaging during the treatment period. Cell death was analyzed by terminal deoxynucleotidyl transferase-mediated dUTP-biotin end labeling of fragmented DNA. RESULTS Tumors can be distinguished with T2-weighted magnetic resonance imaging sequences. Trastuzumab significantly reduced the proliferation of VS cells compared with control (p < 0.01) as analyzed by bromodeoxyuridine uptake. Control tumors demonstrated slight growth during the 12-week treatment period. Both trastuzumab and erlotinib significantly reduced the growth of VS xenografts (p < 0.05). Erlotinib, but not trastuzumab, resulted in a significant increase in the percentage of terminal deoxynucleotidyl transferase-mediated dUTP-biotin end labeling of fragmented DNA-positive VS cells (p < 0.01). CONCLUSION In this preliminary study, the ErbB inhibitors trastuzumab and erlotinib decreased growth of VS xenografts in nude mice, raising the possibility of using ErbB inhibitors in the management of patients with schwannomas, particularly those with neurofibromatosis Type 2.
Collapse
|
31
|
Chen B, Wang Y, Kane SE, Chen S. Improvement of sensitivity to tamoxifen in estrogen receptor-positive and Herceptin-resistant breast cancer cells. J Mol Endocrinol 2008; 41:367-77. [PMID: 18768663 PMCID: PMC3503452 DOI: 10.1677/jme-08-0026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
ERBB2 overexpression in estrogen receptor (ER)-positive breast cancer cells such as BT474 (BT) cells has been found to confer resistance to tamoxifen, and suppression of ERBB2 improves the antiproliferative effects of tamoxifen. In this study, the responsiveness to tamoxifen in the BT/HerR, Herceptin-resistant BT cell lines established through constant Herceptin exposure, was evaluated. Compared with BT cells, improvement of sensitivity to tamoxifen in BT/HerR was demonstrated by ER functional analysis and cell proliferation assay. Tamoxifen in the resistant cell line was found to inhibit 17beta-estradiol-stimulating estrogen-responsive gene pS2 expression more effectively than in BT cells in real-time PCR assay. Western blot analysis showed that cross-phosphorylation between ER and downstream components of ERBB2 was attenuated in BT/HerR cells. ER redistribution from cytoplasm to nucleus could be found in these cells through immunofluorescence and confocal studies, and importantly, chromatin immunoprecipitation studies demonstrated that tamoxifen induced occupancy of the pS2 promoter by ER and nuclear receptor corepressor (NCOR1) instead of coactivator NCOA3 in these cells. Finally, combination of tamoxifen and Herceptin was found to improve the sensitivity of BT/HerR cells to Herceptin. Our results suggest that the ER genomic pathway in the ER-positive and Herceptin-resistant breast cancer cells may be reactivated, allowing tamoxifen therapy to be effective again, and a combination of tamoxifen and Herceptin can be a potential therapeutic strategy for ER-positive and Herceptin-resistant human breast cancer.
Collapse
Affiliation(s)
- Bin Chen
- Department of Surgical Research, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yuanzhong Wang
- Department of Surgical Research, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Susan E. Kane
- Division of Molecular Medicine, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA
| | - Shiuan Chen
- Department of Surgical Research, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
32
|
Sun B, Ranganathan B, Feng SS. Multifunctional poly(d,l-lactide-co-glycolide)/montmorillonite (PLGA/MMT) nanoparticles decorated by Trastuzumab for targeted chemotherapy of breast cancer. Biomaterials 2008; 29:475-86. [DOI: 10.1016/j.biomaterials.2007.09.038] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2007] [Accepted: 09/28/2007] [Indexed: 10/22/2022]
|
33
|
Additive effects of a prolactin receptor antagonist, G129R, and herceptin on inhibition of HER2-overexpressing breast cancer cells. Breast Cancer Res Treat 2007; 111:241-50. [DOI: 10.1007/s10549-007-9789-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Accepted: 10/08/2007] [Indexed: 10/22/2022]
|
34
|
Yang T, Choi MK, Cui FD, Lee SJ, Chung SJ, Shim CK, Kim DD. Antitumor effect of paclitaxel-loaded PEGylated immunoliposomes against human breast cancer cells. Pharm Res 2007; 24:2402-11. [PMID: 17828616 DOI: 10.1007/s11095-007-9425-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 07/31/2007] [Indexed: 11/24/2022]
Abstract
PURPOSE The antitumor effect of paclitaxel-loaded PEGylated immunoliposome (PILs) was investigated in breast cancer cell lines and the xenograft model. METHODS Herceptin was conjugated to paclitaxel-loaded PEGylated liposomes (PLs). In vitro cellular uptake and cytotoxicity of PILs were determined in breast cancer cell lines while in vivo antitumor efficacy was evaluated in the xenograft nude mouse model. RESULTS The PILs formulation was able to significantly increase the HER2 mediated cellular uptake of paclitaxel compared to the PLs in cell lines overexpressing HER2 (BT-474 and SK-BR-3 cells). However, in the MDA-MB-231 cells, which express low levels of HER2, the difference between the PILs and PLs formulation was not significant. The biological activity of Herceptin was maintained throughout the conjugation process as exhibited by the antitumor dose-response curves determined for Herceptin itself, for the thiolated Herceptin alone and subsequently for the immunoliposome-coupled Herceptin. In BT-474 and SK-BR-3 cells, the cytotoxicity of the PILs was more potent than that of Taxol. Moreover, in in vivo studies, PILs showed significantly higher tumor tissue distribution of paclitaxel in the BT-474 xenograft model and more superior antitumor efficacy compared to Taxol and PLs. However, in the MDA-MB-231 xenograft model, PILs and PLs showed similar tumor tissue distribution as well as antitumor activity. CONCLUSIONS These results suggest that HER2-mediated endocytosis is involved in the PILs formulation. The ability of the PILs formulation to efficiently and specifically deliver paclitaxel to the HER2-overexpressing cancer cells implies that it is a promising strategy for tumor-specific therapy for HER2-overexpressing breast cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor
- Chemistry, Pharmaceutical
- Dose-Response Relationship, Drug
- Drug Compounding
- Endocytosis
- Female
- Humans
- Immunoconjugates/chemistry
- Immunoconjugates/metabolism
- Immunoconjugates/pharmacology
- Immunoconjugates/therapeutic use
- Lipids/chemistry
- Liposomes
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Paclitaxel/chemistry
- Paclitaxel/metabolism
- Paclitaxel/pharmacology
- Paclitaxel/therapeutic use
- Polyethylene Glycols/chemistry
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Time Factors
- Tissue Distribution
- Trastuzumab
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Tao Yang
- College of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
35
|
Liu B, Ordonez-Ercan D, Fan Z, Edgerton SM, Yang X, Thor AD. Downregulation of erbB3 abrogates erbB2-mediated tamoxifen resistance in breast cancer cells. Int J Cancer 2007; 120:1874-82. [PMID: 17266042 DOI: 10.1002/ijc.22423] [Citation(s) in RCA: 122] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Receptor tyrosine kinase activity is essential for erbB2 (HER2/neu) promotion of breast carcinogenesis, metastasis and therapeutic resistance. erbB2 kinase can be activated by dimerization with another erbB receptor, most of which bind ligands. Of these, the erbB2/erbB3 heterodimer is the most potent oncogenic complex. erbB2 reportedly requires erbB3 to promote cellular proliferation, although this may occur without changes in erbB2 tyrosine kinase activity in some model systems. Our investigations focus on the role(s) of erbB3 in erbB2-associated kinase activity and tamoxifen resistance. Using tumor-derived cell lines from wild type rat c-neu transgenic mice and human breast cancers, we demonstrate that erbB3 plays a critical role in the activation of erbB2 tyrosine kinase activity and erbB2-associated tumorigenesis. Mechanistically, downregulation of erbB3 by specific siRNA reduces erbB2 tyrosine phosphorylation, decreases the PI-3K/Akt signaling, and inhibits mammary/breast cancer cell proliferation and colony formation. Specific erbB3 siRNA sensitizes erbB2 transfected MCF-7 cells (MCF-7/erbB2) to tamoxifen-associated inhibition of both cell growth and colony formation and enhances tamoxifen-induced apoptosis, in contrast to control siRNA transfected MCF-7/erbB2 cells which are tamoxifen-resistant. Our data indicates that erbB2/erbB3 heterodimerization is a prerequisite for erbB2 tyrosine kinase activation in mammary/breast cancer cells and that downregulation of erbB3 inhibits erbB2-associated procarcinogenic activity via inactivation of the PI-3K/Akt pathway. Furthermore, erbB3 also contributes to erbB2-mediated tamoxifen resistance and therefore may be a clinically relevant therapeutic target in addition to erbB2.
Collapse
Affiliation(s)
- Bolin Liu
- Department of Pathology and College of Medicine, University of Colorado at Denver and Health Sciences Center (UCDHSC), Aurora, CO, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Nesterova M, Johnson N, Cheadle C, Cho-Chung YS. Autoantibody biomarker opens a new gateway for cancer diagnosis. Biochim Biophys Acta Mol Basis Dis 2006; 1762:398-403. [PMID: 16483750 DOI: 10.1016/j.bbadis.2005.12.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2005] [Revised: 12/21/2005] [Accepted: 12/22/2005] [Indexed: 11/26/2022]
Abstract
The list of cancer markers of current interest has grown considerably, but none of the markers used in clinical work is a true tumor marker. These cancer biomarkers are based on the determination of tumor antigens. Here, we report a single method of autoantibody enzyme immunoassay (EIA) screens for a spectrum of serum tumor markers. A comparison of the autoantibody-based EIA to conventional antigen EIA kits, using receiver operating characteristic (ROC) plots, showed that the autoantibody EIA can significantly enhance the sensitivity and specificity of tumor markers. The detection of serum autoantibodies for a spectrum of serum tumor markers, as demonstrated here, suggests that most, if not all, serum cancer biomarkers produce autoantibodies. A unique autoantibody biomarker screening method, as presented here, might therefore facilitate achieving the accurate and early diagnosis of cancer.
Collapse
Affiliation(s)
- M Nesterova
- Cellular Biochemistry Section, Basic Research Laboratory, CCR, National Cancer Institute, Bethesda, MD 20892-1750, USA
| | | | | | | |
Collapse
|