1
|
Markotić A, Omerović J, Marijan S, Režić-Mužinić N, Čikeš Čulić V. Biochemical Pathways Delivering Distinct Glycosphingolipid Patterns in MDA-MB-231 and MCF-7 Breast Cancer Cells. Curr Issues Mol Biol 2024; 46:10200-10217. [PMID: 39329960 PMCID: PMC11430773 DOI: 10.3390/cimb46090608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
The complex structure of glycosphingolipids (GSLs) supports their important role in cell function as modulators of growth factor receptors and glutamine transporters in plasma membranes. The aberrant composition of clustered GSLs within signaling platforms, so-called lipid rafts, inevitably leads to tumorigenesis due to disturbed growth factor signal transduction and excessive uptake of glutamine and other molecules needed for increased energy and structural molecule cell supply. GSLs are also involved in plasma membrane processes such as cell adhesion, and their transition converts cells from epithelial to mesenchymal with features required for cell migration and metastasis. Glutamine activates the mechanistic target of rapamycin complex 1 (mTORC1), resulting in nucleotide synthesis and proliferation. In addition, glutamine contributes to the cancer stem cell GD2 ganglioside-positive phenotype in the triple-negative breast cancer cell line MDA-MB-231. Thieno[2,3-b]pyridine derivative possesses higher cytotoxicity against MDA-MB-231 than against MCF-7 cells and induces a shift to aerobic metabolism and a decrease in S(6)nLc4Cer GSL-positive cancer stem cells in the MDA-MB-231 cell line. In this review, we discuss findings in MDA-MB-231, MCF-7, and other breast cancer cell lines concerning their differences in growth factor receptors and recent knowledge of the main biochemical pathways delivering distinct glycosphingolipid patterns during tumorigenesis and therapy.
Collapse
Affiliation(s)
- Anita Markotić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Jasminka Omerović
- Department of Immunology, University of Split School of Medicine, 21000 Split, Croatia
| | - Sandra Marijan
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Nikolina Režić-Mužinić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| | - Vedrana Čikeš Čulić
- Department of Medical Chemistry and Biochemistry, University of Split School of Medicine, 21000 Split, Croatia
| |
Collapse
|
2
|
Pulido-Capiz A, Chimal-Vega B, Avila-Barrientos LP, Campos-Valenzuela A, Díaz-Molina R, Muñiz-Salazar R, Galindo-Hernández O, García-González V. Auraptene Boosts the Efficacy of the Tamoxifen Metabolites Endoxifen and 4-OH-Tamoxifen in a Chemoresistant ER+ Breast Cancer Model. Pharmaceutics 2024; 16:1179. [PMID: 39339215 PMCID: PMC11435248 DOI: 10.3390/pharmaceutics16091179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
Approximately 80% of breast cancer (BC) cases are estrogen receptor positive (ER+) and sensitive to hormone treatment; Tamoxifen is a prodrug, and its main plasmatic active metabolites are 4-hydroxytamoxifen (4-OH Tam) and endoxifen. Despite the effectiveness of tamoxifen therapy, resistance can be developed. An increment in eukaryotic initiation factor-4A complex (eIF4A) activity can result in tamoxifen-resistant tumor cells. For this work, we developed a cell variant resistant to 4-OH Tam and endoxifen, denominated MCF-7Var E; then, the aim of this research was to reverse the acquired resistance of this variant to tamoxifen metabolites by incorporating the natural compound auraptene. Combination treatments of tamoxifen derivatives and auraptene successfully sensitized the chemoresistant MCF-7Var E. Our data suggest a dual regulation of eIF4A and ER by auraptene. Joint treatments of 4-OH Tam and endoxifen with auraptene identified a novel focus for chemoresistance disruption. Synergy was observed using the auraptene molecule and tamoxifen-derived metabolites, which induced a sensitization in MCF-7Var E cells and ERα parental cells that was not observed in triple-negative breast cancer cells (TNBC). Our results suggest a synergistic effect between auraptene and tamoxifen metabolites in a resistant ER+ breast cancer model, which could represent the first step to achieving a pharmacologic strategy.
Collapse
Affiliation(s)
- Angel Pulido-Capiz
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | | | - Alondra Campos-Valenzuela
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Raúl Díaz-Molina
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Raquel Muñiz-Salazar
- Escuela de Ciencias de la Salud, Universidad Autónoma de Baja California, Campus Ensenada, Ensenada 22890, Mexico;
| | - Octavio Galindo-Hernández
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, Mexico; (A.P.-C.); (B.C.-V.); (A.C.-V.); (R.D.-M.); (O.G.-H.)
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, Mexico
| |
Collapse
|
3
|
O’Neill CE, Sun K, Sundararaman S, Chang JC, Glynn SA. The impact of nitric oxide on HER family post-translational modification and downstream signaling in cancer. Front Physiol 2024; 15:1358850. [PMID: 38601214 PMCID: PMC11004480 DOI: 10.3389/fphys.2024.1358850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/16/2024] [Indexed: 04/12/2024] Open
Abstract
The human epidermal growth factor receptor (HER) family consists of four members, activated by two families of ligands. They are known for mediating cell-cell interactions in organogenesis, and their deregulation has been associated with various cancers, including breast and esophageal cancers. In particular, aberrant epidermal growth factor receptor (EGFR) and HER2 signaling drive disease progression and result in poorer patient outcomes. Nitric oxide (NO) has been proposed as an alternative activator of the HER family and may play a role in this aberrant activation due to its ability to induce s-nitrosation and phosphorylation of the EGFR. This review discusses the potential impact of NO on HER family activation and downstream signaling, along with its role in the efficacy of therapeutics targeting the family.
Collapse
Affiliation(s)
- Ciara E. O’Neill
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| | - Kai Sun
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | | | - Jenny C. Chang
- Houston Methodist Research Institute, Houston, TX, United States
- Dr Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | - Sharon A. Glynn
- Lambe Institute for Translational Research, Discipline of Pathology, School of Medicine, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Sharma T, Zhang Y, Zigrossi A, Cravatt BF, Kastrati I. Dimethyl fumarate inhibits ZNF217 and can be beneficial in a subset of estrogen receptor positive breast cancers. Breast Cancer Res Treat 2023; 201:561-570. [PMID: 37477798 DOI: 10.1007/s10549-023-07037-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
PURPOSE The oncogenic factor ZNF217 promotes aggressive estrogen receptor (ER)+breast cancer disease suggesting that its inhibition may be useful in the clinic. Unfortunately, no direct pharmacological inhibitor is available. Dimethyl fumarate (DMF) exhibits anti-breast cancer activities, in vitro and in pre-clinical in vivo models. Its therapeutic benefits stem from covalent modification of cellular thiols such as protein cysteines, but the full profile of molecular targets mediating its anti-breast cancer effects remains to be determined. METHODS ER+breast cancer cells were treated with DMF followed by cysteine-directed proteomics. Cells with modulated ZNF217 levels were used to probe the efficacy of DMF. RESULTS Covalent modification of ZNF217 by DMF identified by proteomics was confirmed by using a DMF-chemical probe. Inhibition of ZNF217's transcriptional activity by DMF was evident on reported ZNF217-target genes. ZNF217 as an oncogene has been shown to enhance stem-like properties, survival, proliferation, and invasion. Consistent with ZNF217 inhibition, DMF was more effective at blocking these ZNF217-driven phenotypes in cells with elevated ZNF217 expression. Furthermore, partial knockdown of ZNF217 led to a reduction in DMF's efficacy. DMF's in vivo activity was evaluated in a xenograft model of MCF-7 HER2 cells that have elevated expression of ZNF217 and DMF treatment resulted in significant inhibition of tumor growth. CONCLUSION These data indicate that DMF's anti-breast cancer activities in the ER+HER2+models, at least in part, are due to inhibition of ZNF217. DMF is identified as a new covalent inhibitor of ZNF217.
Collapse
Affiliation(s)
- Tanu Sharma
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Yuanjin Zhang
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Alexandra Zigrossi
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA
| | - Benjamin F Cravatt
- The Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Irida Kastrati
- The Department of Cancer Biology, Loyola University Chicago, 2160 S 1St Avenue, Maywood, IL, 60153, USA.
| |
Collapse
|
5
|
Type of adjuvant endocrine therapy and disease-free survival in patients with early HR-positive/HER2-positive BC: analysis from the phase III randomized ShortHER trial. NPJ Breast Cancer 2023; 9:6. [PMID: 36739285 PMCID: PMC9899279 DOI: 10.1038/s41523-023-00509-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/21/2023] [Indexed: 02/05/2023] Open
Abstract
The optimal adjuvant endocrine therapy for HR-positive/HER2-positive breast cancer patients is unknown. We included in this analysis 784 patients with HR-positive/HER2-positive BC from the randomized ShortHER trial of adjuvant trastuzumab (1 year vs 9 weeks) + chemotherapy. At a median follow-up of 8.7 years, patients who received AI had a significantly better DFS vs patients who received TAM or TAM-AI: 8-yr DFS 86.4 vs 79.7%, log-rank P = 0.013 (HR 1.52, 95% CI 1.09-2.11). In multivariate analysis, the type of endocrine therapy maintained a significant association with DFS (HR 1.64, 95% CI 1.07-2.52, p = 0.025 for TAM/TAM-AI vs AI). Among premenopausal patients aged ≤45 years, the use of GnRHa was associated with longer DFS: 8-yr DFS rate 85.2 vs 62.6% (log-rank p = 0.019, HR 0.41, 95% CI 0.19-0.88). In this post-hoc analysis of the ShortHER trial adjuvant treatment with AI was independently associated with improved DFS. Subgroup analysis in premenopausal patients suggests benefits with ovarian suppression.Trial registration: NCI ClinicalTrials.gov number: NCT00629278.
Collapse
|
6
|
Effects of Combined Pentadecanoic Acid and Tamoxifen Treatment on Tamoxifen Resistance in MCF−7/SC Breast Cancer Cells. Int J Mol Sci 2022; 23:ijms231911340. [PMID: 36232636 PMCID: PMC9570034 DOI: 10.3390/ijms231911340] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Estrogen receptors are indicators of breast cancer adaptability to endocrine therapies, such as tamoxifen. Deficiency or absence of estrogen receptor α (ER−α) in breast cancer cells results in reduced efficacy of endocrine therapy. Here, we investigated the effect of combined tamoxifen and pentadecanoic acid therapy on ER−α−under−expressing breast cancer cells. Drug resistance gene expression patterns were determined by RNA sequencing analysis and in vitro experiments. For the first time, we demonstrate that the combined treatment of pentadecanoic acid, an odd−chain fatty acid, and tamoxifen synergistically suppresses the growth of human breast carcinoma MCF−7 stem cells (MCF−7/SCs), which were found to be tamoxifen−resistant and showed reduced ER−α expression compared with the parental MCF−7 cells. In addition, the combined treatment synergistically induced apoptosis and accumulation of sub−G1 cells and suppressed epithelial−to−mesenchymal transition (EMT). Exposure to this combination induces re−expression of ER−α at the transcriptional and protein levels, along with suppression of critical survival signal pathways, such as ERK1/2, MAPK, EGFR, and mTOR. Collectively, decreased ER−α expression was restored by pentadecanoic acid treatment, resulting in reversal of tamoxifen resistance. Overall, pentadecanoic acid exhibits the potential to enhance the efficacy of endocrine therapy in the treatment of ER−α−under−expressing breast cancer cells.
Collapse
|
7
|
Saleh AB, Hassan NH, Ismail MA, El-Sayed WM. New fluorobenzamidine exerts antitumor activity against breast cancer in mice via pro-apoptotic activity. Discov Oncol 2022; 13:88. [PMID: 36107265 PMCID: PMC9478011 DOI: 10.1007/s12672-022-00554-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 08/30/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Breast cancer is one of the leading causes of cancer-related morbidities. The present study aimed to evaluate the efficacy of bithiophene-fluorobenzamidine (BFB) against breast cancer induced by 7,12-dimethylbenz(a)anthracene (DMBA) in female Swiss mice and reveal the underlining mechanisms. METHODS The mice were randomly divided into five groups; control, BFB-treated group, DMBA-treated group, and the last two groups received DMBA then tamoxifen or BFB. RESULTS BFB reduced the tumor incidence by ~ 88% versus 30% after TAM. DMBA significantly increased the expression of CDK1 and HER2 and reduced the expression of p53, p21 (CDKN1A), ESR-α, and CAS3. BFB caused significant down-regulation of CDK1 and HER2 and upregulation of p53, p21, ESR-α, and CAS3. In the DMBA-treated mice, cancerous cells metastasized to several organs. This was prevented by the administration of BFB. The antimetastatic and proapoptotic activities were confirmed in MCF7 cells in vitro by the wound healing and annexin V assays, respectively. Kaplan-Meier analysis showed that the BFB increased survival. In the DMBA group, tumors showed invasive carcinoma of grade III with central necrosis, polymorphism, mitotic activity, and numerous newly formed ductules, and colloidal mucinous secretions within adenoid cysts. BFB administration restored the normal structure of the mammary glands. CONCLUSION Taken together, BFB has antitumor, pro-apoptotic, and anti-metastatic activities against breast cancer in mice and therefore, it merits further investigations.
Collapse
Affiliation(s)
- AbdelRahman B Saleh
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Nagwa H Hassan
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt
| | - Mohamed A Ismail
- Department of Chemistry, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| | - Wael M El-Sayed
- Department of Zoology, Faculty of Science, Ain Shams University, Cairo, 11566, Egypt.
| |
Collapse
|
8
|
Alataki A, Dowsett M. Human epidermal growth factor receptor-2 and endocrine resistance in hormone-dependent breast cancer. Endocr Relat Cancer 2022; 29:R105-R122. [PMID: 35613334 PMCID: PMC9254309 DOI: 10.1530/erc-21-0293] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 12/27/2022]
Abstract
Endocrine therapies are the main treatment strategies for the clinical management of hormone-dependent breast cancer. Despite prolonged time to recurrence in the adjuvant setting and the initial clinical responses in the metastatic setting, many patients eventually encounter tumour relapse due to acquired resistance to these agents. Other patients experience a lack of tumour regression at the beginning of treatment indicating de novo resistance that significantly limits its efficacy in the clinic. There is compelling evidence that human epidermal growth factor receptor-2 (HER2) overexpression contributes to resistance to endocrine therapies in oestrogen receptor-positive (ER+) breast cancer. ER+/HER2+ tumours comprise about 10% of all breast cancer cases and about 60% of the whole set of HER2+ tumours. Most patients with primary ER+/HER2+ disease will receive antibody-based HER2-targeted therapy, but this is generally for no more than one year while endocrine treatment is usually for at least 5 years. A number of HER2-kinase inhibitors are also now in clinical use or in clinical trials, and the interaction of these with endocrine treatment may differ from that of antibody treatment. In this review article, we aim to summarise knowledge on molecular mechanisms of breast cancer resistance to endocrine therapies attributable to the impact of HER2 signalling on endocrine sensitivity, to discuss data from clinical trials addressing the role of HER2 in the development of endocrine resistance in the metastatic, neoadjuvant and adjuvant settings and to explore rational new therapeutic strategies.
Collapse
Affiliation(s)
- Anastasia Alataki
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital and The Institute of Cancer Research, London, UK
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Correspondence should be addressed to A Alataki:
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital and The Institute of Cancer Research, London, UK
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| |
Collapse
|
9
|
Huang L, Liu X, Li L, Wang L, Wu N, Liu Z. Novel immune subtypes identification of HER2-positive breast cancer based on immunogenomic landscape. Med Oncol 2022; 39:92. [PMID: 35568771 DOI: 10.1007/s12032-022-01690-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
HER2 positive BC is heterogeneous. But few studies discussed the classification of HER2-positive BC based on immune-related signatures. Using three publicly BC genomics datasets, we classified HER2 positive BC based on 33 immune-related signatures and used unsupervised machine learning methods to predict and perform the classification. We grouped three HER2-positive BC subtypes that we called Immune-High (IM-H), Immune-Medium (IM-M), and Immune-Low (IM-L), and manifested this categorization was predictable, duplicable and reliable by analyzing another dataset. Compared to other subtypes, IM-H had a higher immune cell infiltration level and stronger anti-tumor immune activities, as well as better clinical survival outcome. Besides these signatures, there were some cancer-related pathways which were hyperactivated in IM-H, including cytokine-cytokine receptor interactions, antigen processing and presentation pathways, natural killer cell-mediated cytotoxicity, Th1 and Th2 cell differentiation, chemokine signaling pathway, Th17 cell differentiation, B and T cell receptor signaling, NF-kappa B signaling, PD-L1 expression and PD-1 checkpoint pathway in cancer, TNF signaling, IL-17 signaling, NOD-like receptor signaling and Toll-like receptor signaling. By contrast, IM-L showed depressed immune-related signatures and enhanced activation of lycosylphosphatidylinositol-anchor biosynthesis and mismatch repair. Moreover, we discovered a gene co-expression network focused on eight transcription factor genes (EOMES, TBX21, GFI1, IRF4, POU2AF1, CIITA, FOXP3 and TOX) and one tumor suppress gene (PRF1), which were closely related with tumor immune. We identified three HER2-positive BC subtypes based on immune-related signatures, which had potential clinical implications and promoted the optimal stratification of HER2-positive BC responsive to immunotherapy.
Collapse
Affiliation(s)
- Lingli Huang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Xin Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Li Li
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Lei Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Nan Wu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China
| | - Zhixian Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
10
|
Chen M, Sheu MT, Cheng TL, Roffler SR, Lin SY, Chen YJ, Cheng YA, Cheng JJ, Chang HY, Wu TY, Kao AP, Ho YS, Chuang KH. A novel anti-tumor/anti-tumor-associated fibroblast/anti-mPEG tri-specific antibody to maximize the efficacy of mPEGylated nanomedicines against fibroblast-rich solid tumor. Biomater Sci 2021; 10:202-215. [PMID: 34826322 DOI: 10.1039/d1bm01218e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The therapeutic efficacy of methoxypolyethylene glycol (mPEG)-coated nanomedicines in solid tumor treatment is hindered by tumor-associated fibroblasts (TAFs), which promote tumor progression and form physical barriers. We developed an anti-HER2/anti-FAP/anti-mPEG tri-specific antibody (TsAb) for one-step conversion of mPEG-coated liposomal doxorubicin (Lipo-Dox) to immunoliposomes, which simultaneously target HER2+ breast cancer cells and FAP+ TAFs. The non-covalent modification did not adversely alter the physical characteristics and stability of Lipo-Dox. The TsAb-Lipo-Dox exhibited specific targeting and enhanced cytotoxicity against mono- and co-cultured HER2+ breast cancer cells and FAP+ TAFs, compared to bi-specific antibody (BsAb) modified or unmodified Lipo-Dox. An in vivo model of human breast tumor containing TAFs also revealed the improved tumor accumulation and therapeutic efficacy of TsAb-modified mPEGylated liposomes without signs of toxicity. Our data indicate that arming clinical mPEGylated nanomedicines with the TsAb is a feasible and applicable approach for overcoming the difficulties caused by TAFs in solid tumor treatment.
Collapse
Affiliation(s)
- Michael Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Ming-Thau Sheu
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tian-Lu Cheng
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Steve R Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Shyr-Yi Lin
- Department of Primary Care Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jou Chen
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yi-An Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung City, Taiwan
| | - Jing-Jy Cheng
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan
| | - Hsin-Yu Chang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Tung-Yun Wu
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan
| | - An-Pei Kao
- Stemforce Biotechnology Co., Ltd, Chiayi City, Taiwan
| | - Yuan-Soon Ho
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.,School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,Cancer Research Center, Taipei Medical University Hospital, Taipei, Taiwan
| | - Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan.,Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.,Ph.D. Program in Clinical Drug Development of Herbal Medicine, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei, Taiwan. .,Ph.D Program in Biotechnology Research and Development, Taipei Medical University, Taipei, Taiwan.,Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
11
|
Hurvitz SA, McAndrew NP, Bardia A, Press MF, Pegram M, Crown JP, Fasching PA, Ejlertsen B, Yang EH, Glaspy JA, Slamon DJ. A careful reassessment of anthracycline use in curable breast cancer. NPJ Breast Cancer 2021; 7:134. [PMID: 34625570 PMCID: PMC8501074 DOI: 10.1038/s41523-021-00342-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
It has been over three decades since anthracyclines took their place as the standard chemotherapy backbone for breast cancer in the curative setting. Though the efficacy of anthracycline chemotherapy is not debatable, potentially life-threatening and long-term risks accompany this class of agents, leading some to question their widespread use, especially when newer agents with improved therapeutic indices have become available. Critically assessing when to incorporate an anthracycline is made more relevant in an era where molecular classification is enabling not only the development of biologically targeted therapeutics but also is improving the ability to better select those who would benefit from cytotoxic agents. This comprehensive analysis will present the problem of overtreatment in early-stage breast cancer, review evidence supporting the use of anthracyclines in the pre-taxane era, analyze comparative trials evaluating taxanes with or without anthracyclines in biologically unselected and selected patient populations, and explore published work aimed at defining anthracycline-sensitive tumor types.
Collapse
Affiliation(s)
- Sara Alsterlind Hurvitz
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Nicholas P. McAndrew
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Aditya Bardia
- grid.38142.3c000000041936754XMassachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Michael F. Press
- grid.42505.360000 0001 2156 6853University of Southern California, Los Angeles, CA USA
| | - Mark Pegram
- Stanford Comprehensive Cancer Institute, Palo Alto, CA USA
| | - John P. Crown
- grid.412751.40000 0001 0315 8143Department of Medical Oncology, St. Vincent’s University Hospital, Dublin, Ireland
| | - Peter A. Fasching
- grid.411668.c0000 0000 9935 6525Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Bent Ejlertsen
- grid.4973.90000 0004 0646 7373Department of Oncology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Eric H. Yang
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - John A. Glaspy
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| | - Dennis J. Slamon
- grid.19006.3e0000 0000 9632 6718Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Jonsson Comprehensive Cancer Center, Los Angeles, CA USA
| |
Collapse
|
12
|
Byrd BK, Strawbridge RS, Wells W, Barth C, Gibbs S, Davis SC. A method for validating depth-resolved biodistributions in topically-stained specimen with multi-channel fluorescence cryo-imaging. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2021; 11625. [PMID: 34475612 DOI: 10.1117/12.2582542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Fluorescent contrast agents targeted to cancer biomarkers are increasingly being explored for cancer detection, surgical guidance, and response monitoring. Efforts have been underway to topically apply such biomarker-targeted agents to freshly excised specimen for detecting cancer cell receptors on the surface as a method for intraoperative surgical margin assessment, including dual-probe staining methods introduce a second 'non-specific' optical agent as a control to help compensate for heterogeneous uptake and normalize the imaging field. Still, such specimen staining protocols introduce multifaceted complexity with unknown variables, such as tissue-specific diffusion, cell-specific binding and disassociation rates, and other factors, affecting the interpreted cancer-biomarker distribution across the specimen surface. The ability to recover three-dimensional dual-probe biodistributions throughout whole-specimens could offer a ground-truth validation method for examining topical staining uptake behaviors. Herein, we report on a novel method for characterizing dual-probe accumulation with 3D depth-profiles observed from a dual-probe fresh-specimen staining experiment.
Collapse
Affiliation(s)
- Brook K Byrd
- Dartmouth College, Thayer School of Engineering, Hanover, NH, 03755
| | | | - Wendy Wells
- Dartmouth Hitchcock Medical Center, Department of Surgery, Lebanon, NH, 03766
| | - Connor Barth
- Oregon Heath and Sciences University, Portland, OR, 97239
| | - Summer Gibbs
- Oregon Heath and Sciences University, Portland, OR, 97239
| | - Scott C Davis
- Dartmouth College, Thayer School of Engineering, Hanover, NH, 03755
| |
Collapse
|
13
|
Hurvitz SA. Anthracycline Use in ERBB2-Positive Breast Cancer: It Is Time to Re-TRAIN. JAMA Oncol 2021; 7:975-977. [PMID: 34014297 DOI: 10.1001/jamaoncol.2021.1314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Sara A Hurvitz
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles
| |
Collapse
|
14
|
Fatty Acid Synthase Confers Tamoxifen Resistance to ER+/HER2+ Breast Cancer. Cancers (Basel) 2021; 13:cancers13051132. [PMID: 33800852 PMCID: PMC7961649 DOI: 10.3390/cancers13051132] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary Overactivation of the human epidermal growth factor receptor 2 (HER2) is one of the main drivers of tamoxifen resistance in estrogen receptor (ER)-positive breast cancer patients. Combined targeting of HER2 and ER, however, has yielded disappointing results in the clinical setting. Therefore, other potential mechanisms for tamoxifen resistance would not be overcome by solely blocking the cross-talk between ER and HER2 at the receptor(s) level. Using cell lines, animal models, and clinical data, we provide evidence to support a critical role of fatty acid synthase (FASN)—the major site for endogenous fat synthesis—in HER2-driven tamoxifen resistance. Importantly, treatment with a FASN inhibitor impeded the estrogen-like tumor-promoting effects of tamoxifen and fully restored the anti-estrogenic activity of tamoxifen in ER+/HER2-overexpressing breast cancer xenografts. We postulate FASN as a biological determinant of HER2-driven tamoxifen resistance and FASN inhibition as a novel therapeutic approach to restore tamoxifen sensitivity in endocrine-resistant breast cancer. Abstract The identification of clinically important molecular mechanisms driving endocrine resistance is a priority in estrogen receptor-positive (ER+) breast cancer. Although both genomic and non-genomic cross-talk between the ER and growth factor receptors such as human epidermal growth factor receptor 2 (HER2) has frequently been associated with both experimental and clinical endocrine therapy resistance, combined targeting of ER and HER2 has failed to improve overall survival in endocrine non-responsive disease. Herein, we questioned the role of fatty acid synthase (FASN), a lipogenic enzyme linked to HER2-driven breast cancer aggressiveness, in the development and maintenance of hormone-independent growth and resistance to anti-estrogens in ER/HER2-positive (ER+/HER2+) breast cancer. The stimulatory effects of estradiol on FASN gene promoter activity and protein expression were blunted by anti-estrogens in endocrine-responsive breast cancer cells. Conversely, an AKT/MAPK-related constitutive hyperactivation of FASN gene promoter activity was unaltered in response to estradiol in non-endocrine responsive ER+/HER2+ breast cancer cells, and could be further enhanced by tamoxifen. Pharmacological blockade with structurally and mechanistically unrelated FASN inhibitors fully impeded the strong stimulatory activity of tamoxifen on the soft-agar colony forming capacity—an in vitro metric of tumorigenicity—of ER+/HER2+ breast cancer cells. In vivo treatment with a FASN inhibitor completely prevented the agonistic tumor-promoting activity of tamoxifen and fully restored its estrogen antagonist properties against ER/HER2-positive xenograft tumors in mice. Functional cancer proteomic data from The Cancer Proteome Atlas (TCPA) revealed that the ER+/HER2+ subtype was the highest FASN protein expressor compared to basal-like, HER2-enriched, and ER+/HER2-negative breast cancer groups. FASN is a biological determinant of HER2-driven endocrine resistance in ER+ breast cancer. Next-generation, clinical-grade FASN inhibitors may be therapeutically relevant to countering resistance to tamoxifen in FASN-overexpressing ER+/HER2+ breast carcinomas.
Collapse
|
15
|
Skok K, Gradišnik L, Maver U, Kozar N, Sobočan M, Takač I, Arko D, Kavalar R. Gynaecological cancers and their cell lines. J Cell Mol Med 2021; 25:3680-3698. [PMID: 33650759 PMCID: PMC8051715 DOI: 10.1111/jcmm.16397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/24/2022] Open
Abstract
Cell lines are widely used for various research purposes including cancer and drug research. Recently, there have been studies that pointed to discrepancies in the literature and usage of cell lines. That is why we have prepared a comprehensive overview of the most common gynaecological cancer cell lines, their literature, a list of currently available cell lines, and new findings compared with the original studies. A literature review was conducted via MEDLINE, PubMed and ScienceDirect for reviews in the last 5 years to identify research and other studies related to gynaecological cancer cell lines. We present an overview of the current literature with reference to the original studies and pointed to certain inconsistencies in the literature. The adherence to culturing rulesets and the international guidelines helps in minimizing replication failure between institutions. Evidence from the latest research suggests that despite certain drawbacks, variations of cancer cell lines can also be useful in regard to a more diverse genomic landscape.
Collapse
Affiliation(s)
- Kristijan Skok
- Department of pathology, General Hospital Graz II, Graz, Austria.,Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Lidija Gradišnik
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Nejc Kozar
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Monika Sobočan
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Iztok Takač
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Darja Arko
- Division of Gynecology and Perinatology, University Medical Center Maribor, Maribor, Slovenia.,Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rajko Kavalar
- Faculty of Medicine, University of Maribor, Maribor, Slovenia.,Department of Pathology, University Medical Center Maribor, Maribor, Slovenia
| |
Collapse
|
16
|
Blatt EB, Kopplin N, Kumar S, Mu P, Conzen SD, Raj GV. Overcoming oncogene addiction in breast and prostate cancers: a comparative mechanistic overview. Endocr Relat Cancer 2021; 28:R31-R46. [PMID: 33263560 PMCID: PMC8218927 DOI: 10.1530/erc-20-0272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
Prostate cancer (PCa) and breast cancer (BCa) are both hormone-dependent cancers that require the androgen receptor (AR) and estrogen receptor (ER, ESR1) for growth and proliferation, respectively. Endocrine therapies that target these nuclear receptors (NRs) provide significant clinical benefit for metastatic patients. However, these therapeutic strategies are seldom curative and therapy resistance is prevalent. Because the vast majority of therapy-resistant PCa and BCa remain dependent on the augmented activity of their primary NR driver, common mechanisms of resistance involve enhanced NR signaling through overexpression, mutation, or alternative splicing of the receptor, coregulator alterations, and increased intracrine hormonal synthesis. In addition, a significant subset of endocrine therapy-resistant tumors become independent of their primary NR and switch to alternative NR or transcriptional drivers. While these hormone-dependent cancers generally employ similar mechanisms of endocrine therapy resistance, distinct differences between the two tumor types have been observed. In this review, we compare and contrast the most frequent mechanisms of antiandrogen and antiestrogen resistance, and provide potential therapeutic strategies for targeting both advanced PCa and BCa.
Collapse
Affiliation(s)
- Eliot B Blatt
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Noa Kopplin
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shourya Kumar
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ping Mu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suzanne D Conzen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ganesh V Raj
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
17
|
Hormati A, Shiran JA, Molazadeh M, Kaboudin B, Ahmadpour S. Synthesis of New Thioureas Derivatives and Evaluation of Their Efficacy as Proliferation Inhibitors in MCF-7 Breast Cancer Cells by Using 99mTc-MIBI Radiotracer. Med Chem 2021; 17:766-778. [PMID: 32334505 DOI: 10.2174/1573406416666200425224921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/26/2020] [Accepted: 03/26/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Anti-tumor activity of some thioureas derivatives is well documented in literature and received considerable attention. The present study aims to synthesize and characterize some novel thioureas and carbonylthioureas as anti-tumor agents for MCF-7 breast cancer cells in vitro and in vivo. MATERIALS AND METHODS Several 1-allyl-3-(substituted phenyl), N,N'-(phenylene) bis(3- allyldithithiourea) and 1-cyclopropanecarbonyl-3-(substituted phenyl)-thioureas derivatives were synthesized and confirmed by FT-IR spectroscopy, NMR and 13C-NMR. Anti-tumor activity of these compounds was determined by various in vitro and in vivo assays including; MTT, tumor volume measurement as well as,99mTc-MIBI tumor uptake in MCF-7 tumor bearing nude mice. RESULTS Among all of the synthesized compounds, some thioureas derivatives [3i] and [4b] at 100 nM concentration exhibited significant inhibitory effects on the proliferation of MCF-7 cell in vitro. However, this inhibition was not observed in HUVEC human endothelial normal cells. In vivo anti-tumor effects of the synthesized compounds on MCF-7 xenograft mouse models demonstrated a reduction in the tumor volume for various concentrations between 2 to 10 mg/kg after 21 days. These effects were comparable with Tamoxifen as standard anti-estrogen drug. According to the 99mTc-MIBI biodistribution result, treatment of MCF-7 bearing nude mice with both [3i] and [4b] compounds at the maximum concentration (10 mg/kg) can lead to a significant decrease of 99mTc- MIBI tumor uptake. CONCLUSION Compounds [3i] and [4b] suppressed the growth of MCF-7 cells in the xenograft nude mice at the doses that were well-tolerated. Our study suggests that these new compounds with their significant anti-tumor effects, may serve as useful candidates for breast cancer therapy.
Collapse
Affiliation(s)
- Ahmad Hormati
- Gastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Jafar Abbasi Shiran
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Mikaeil Molazadeh
- Department of Medical Physics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Babak Kaboudin
- Department of Chemistry, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Sajjad Ahmadpour
- Gastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
18
|
Ammazzalorso A, Gallorini M, Fantacuzzi M, Gambacorta N, De Filippis B, Giampietro L, Maccallini C, Nicolotti O, Cataldi A, Amoroso R. Design, synthesis and biological evaluation of imidazole and triazole-based carbamates as novel aromatase inhibitors. Eur J Med Chem 2020; 211:113115. [PMID: 33360796 DOI: 10.1016/j.ejmech.2020.113115] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/07/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022]
Abstract
In the search for novel aromatase inhibitors, a series of triazole and imidazole-based carbamate derivatives were designed and synthesized. Final compounds were thus evaluated against human aromatase by in vitro kinetic experiments in a fluorimetric assay in comparison with letrozole. The effect of most active derivatives 13a and 15c was then evaluated in vitro on the human breast cancer cell line MCF7 by MTT assay, cytotoxicity assay (LDH release) and cell cycle analysis, revealing a dose-dependent inhibition profile of cell viability and low micromolar IC50 values. In addition, docking simulations were also carried out to elucidate at a molecular level of detail the binding modes adopted to target human aromatase.
Collapse
Affiliation(s)
- Alessandra Ammazzalorso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy.
| | - Marialucia Gallorini
- Unit of Anatomy, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Marialuigia Fantacuzzi
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Nicola Gambacorta
- Unit of Medicinal Chemistry, Department of Farmacia-Scienze Del Farmaco, "A. Moro" University, Bari, Italy
| | - Barbara De Filippis
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Letizia Giampietro
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Cristina Maccallini
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Orazio Nicolotti
- Unit of Medicinal Chemistry, Department of Farmacia-Scienze Del Farmaco, "A. Moro" University, Bari, Italy
| | - Amelia Cataldi
- Unit of Anatomy, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| | - Rosa Amoroso
- Unit of Medicinal Chemistry, Department of Pharmacy, "G. D'Annunzio" University, Chieti, Italy
| |
Collapse
|
19
|
Shafran Y, Deutsch M, Afrimzon E, Ravid-Hermesh O, Sobolev M, Bar-On-Eizig Z, Shainberg A, Zurgil N. Co-culture hydrogel micro-chamber array-based plate for anti-tumor drug development at single-element resolution. Toxicol In Vitro 2020; 71:105067. [PMID: 33301902 DOI: 10.1016/j.tiv.2020.105067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/19/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022]
Abstract
In response to the need for reliable cellular models that reflect complex tumor microenvironmental properties, and enable more precise testing of anti-cancer therapeutics effects on humans, a co-culture platform for in-vitro model that enhances the physiology of breast cancer (BC) microenvironment is presented. A six well imaging plate wherein each macro-well contains several separate compartments was designed. Three-dimensional (3D) cancer spheroids are generated and cultured in the inner compartment which is embossed with an array of nano-liter micro-chambers made of hydrogel. Stromal cells are cultured in the outer chambers. The two cell types are cultured side-by-side, sharing a common space, thus enabling extra-cellular communication via secreted molecules. As proof of concept, a model of BC tumor microenvironment was recapitulated by co-cultivating 3D MCF7 spheroids in the presence of tumor-associated macrophages (TAMs). The presence of TAMs induced an aggressive phenotype by promoting spheroid growth, enhancing survivin expression levels and enabling invasive behavior. Moreover, TAMs influenced the response of BC spheroids to cytotoxic treatment as well as hormonal drug therapy, and enhanced the effects of nitric oxide donor. The platform enables time-lapse imaging and treatment without losing spatial location of the measured spheroids, thereby allowing measurements and analysis at individual-object resolution in an easy and efficient manner.
Collapse
Affiliation(s)
- Yana Shafran
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Mordechai Deutsch
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Elena Afrimzon
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Orit Ravid-Hermesh
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Maria Sobolev
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Zehavit Bar-On-Eizig
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Asher Shainberg
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Naomi Zurgil
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar-Ilan University, Ramat-Gan 52900, Israel.
| |
Collapse
|
20
|
A novel cancer preventative botanical mixture, TriCurin, inhibits viral transcripts and the growth of W12 cervical cells harbouring extrachromosomal or integrated HPV16 DNA. Br J Cancer 2020; 124:901-913. [PMID: 33257842 PMCID: PMC7921087 DOI: 10.1038/s41416-020-01170-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The phytochemical mixture TriCurin (curcumin, epigallocatechin gallate (EGCG) and resveratrol) eliminates human papillomavirus (HPV) (+) cancer cells in vitro and in vivo. In this study, we further evaluate TriCurin. METHODS The activity of TriCurin and its individual compounds was assayed on W12 cells, derived from a cervical precancer containing episomal and integrated HPV16 DNA, using MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assays, microscopy and reverse transcription-polymerase chain reaction (RT-PCR), and on HeLa cells by gene expression analysis. The stability and toxicity of TriCurin microemulsion were tested in an organotypic cervical tissue model. RESULTS TriCurin and its individual compounds inhibit the growth of W12 cells, episomal, type 1 and 2 integrants; the relative order of activity is TriCurin, EGCG, curcumin, or resveratrol. RT-PCR shows that TriCurin activates p53 and suppresses HPV16 mRNAs E1, E2, E4, E6 and E7 at 24 h in W12 cells. Gene expression analysis shows that TriCurin activates pro-apoptotic genes and represses anti-apoptotic genes in HeLa cells. TriCurin in a microemulsion is stable and non-toxic to cervical tissue. The combination of TriCurin and tanshinone IIA exhibits additional synergy against HeLa cells. CONCLUSIONS TriCurin, and the combination of TriCurin with tanshinone IIA, are effective against HPV (+) cells. The phytochemical mixture, in the microemulsion-based cream, is a promising therapeutic for the prevention and treatment of cervical cancer.
Collapse
|
21
|
Song PN, Mansur A, Dugger KJ, Davis TR, Howard G, Yankeelov TE, Sorace AG. CD4 T-cell immune stimulation of HER2 + breast cancer cells alters response to trastuzumab in vitro. Cancer Cell Int 2020; 20:544. [PMID: 33292267 PMCID: PMC7654187 DOI: 10.1186/s12935-020-01625-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction The HER2 + tumor immune microenvironment is composed of macrophages, natural killer cells, and tumor infiltrating lymphocytes, which produce pro-inflammatory cytokines. Determining the effect of T-cells on HER2 + cancer cells during therapy could guide immunogenic therapies that trigger antibody-dependent cellular cytotoxicity. This study utilized longitudinal in vitro time-resolved microscopy to measure T-cell influence on trastuzumab in HER2 + breast cancer. Methods Fluorescently-labeled breast cancer cells (BT474, SKBR3, MDA-MB-453, and MDA-MB-231) were co-cultured with CD4 + T-cells (Jurkat cell line) and longitudinally imaged to quantify cancer cell viability when treated with or without trastuzumab (10, 25, 50 and 100 μg/mL). The presence and timing of T-cell co-culturing was manipulated to determine immune stimulation of trastuzumab-treated HER2 + breast cancer. HER2 and TNF-α expression were evaluated with western blot and ELISA, respectively. Significance was calculated using a two-tailed parametric t-test. Results The viability of HER2 + cancer cells significantly decreased when exposed to 25 μg/mL trastuzumab and T-cells, compared to cancer cells exposed to trastuzumab without T-cells (p = 0.01). The presence of T-cells significantly increased TNF-α expression in trastuzumab-treated cancer cells (p = 0.02). Conversely, cancer cells treated with TNF-α and trastuzumab had a similar decrease in viability as trastuzumab-treated cancer cells co-cultured with T-cells (p = 0.32). Conclusions The presence of T-cells significantly increases the efficacy of targeted therapies and suggests trastuzumab may trigger immune mediated cytotoxicity. Increased TNF-α receptor expression suggest cytokines may interact with trastuzumab to create a state of enhanced response to therapy in HER2 + breast cancer, which has potential to reducing tumor burden.
Collapse
Affiliation(s)
- Patrick N Song
- Department of Radiology, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, 35233, USA
| | - Ameer Mansur
- Department of Biomedical Engineering, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, 35233, USA
| | - Kari J Dugger
- Department of Clinical and Diagnostic Sciences, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tessa R Davis
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Grant Howard
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Thomas E Yankeelov
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.,Department of Diagnostic Medicine, The University of Texas at Austin, Austin, TX, USA.,Department of Oncology, The University of Texas at Austin, Austin, TX, USA.,Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX, USA.,Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX, USA
| | - Anna G Sorace
- Department of Radiology, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, 35233, USA. .,Department of Biomedical Engineering, The University of Alabama at Birmingham, 1670 University Blvd, Birmingham, AL, 35233, USA. .,O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
22
|
Solomon VR, Alizadeh E, Bernhard W, Makhlouf A, Hartimath SV, Hill W, El-Sayed A, Barreto K, Geyer CR, Fonge H. Development and preclinical evaluation of cixutumumab drug conjugates in a model of insulin growth factor receptor I (IGF-1R) positive cancer. Sci Rep 2020; 10:18549. [PMID: 33122707 PMCID: PMC7596529 DOI: 10.1038/s41598-020-75279-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 09/29/2020] [Indexed: 12/25/2022] Open
Abstract
Overexpression of insulin growth factor receptor type 1 (IGF-1R) is observed in many cancers. Antibody drug conjugates (ADCs) with PEGylated maytansine (PEG6-DM1) show promise in vitro. We developed PEG6-DM1 ADCs with low and high drug to antibody ratios (DAR) using an anti-IGF-1R antibody cixutumumab (IMC-A12). Conjugates with low (cixutumumab-PEG6-DM1-Low) and high (cixutumumab-PEG6-DM1-High) DAR as 3.4 and 7.2, respectively, were generated. QC was performed by UV spectrophotometry, HPLC, bioanalyzer, and biolayer-interferometry. We compared the in vitro binding and internalization rates of the ADCs in IGF-1R-positive MCF-7/Her18 cells. We radiolabeled the ADCs with 111In and used microSPECT/CT imaging and ex vivo biodistribution to understand their in vivo behavior in MCF-7/Her18 xenograft mice. The therapeutic potential of the ADC was studied in vitro and in mouse xenograft. Internalization rates of all ADCs was high and increased over 48 h and EC50 was in the low nanomolar range. MicroSPECT/CT imaging and ex vivo biodistribution showed significantly lower tumor uptake of 111In-cixutumumab-PEG6-DM1-High compared to 111In-cixutumumab-PEG6-DM1-Low and 111In-cixutumumab. Cixutumumab-PEG6-DM1-Low significantly prolonged the survival of mice bearing MCF-7/Her18 xenograft compared with cixutumumab, cixutumumab-PEG6-DM1-High, or the PBS control group. Cixutumumab-PEG6-DM1-Low ADC was more effective. The study highlights the potential utility of cixutumumab-ADCs as theranostics against IGF-1R positive cancers.
Collapse
Affiliation(s)
- Viswas Raja Solomon
- Department of Medical Imaging, RUH Saskatoon, University of Saskatchewan, College of Medicine, 103 Hospital Dr., Saskatoon, SK, S7N 0W8, Canada
| | - Elahe Alizadeh
- Department of Medical Imaging, RUH Saskatoon, University of Saskatchewan, College of Medicine, 103 Hospital Dr., Saskatoon, SK, S7N 0W8, Canada
| | - Wendy Bernhard
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, College of Medicine, Saskatoon, SK, Canada
| | - Amal Makhlouf
- Department of Medical Imaging, RUH Saskatoon, University of Saskatchewan, College of Medicine, 103 Hospital Dr., Saskatoon, SK, S7N 0W8, Canada.,Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Kasr El-Aini, Cairo, 12411, Egypt
| | - Siddesh V Hartimath
- Department of Medical Imaging, RUH Saskatoon, University of Saskatchewan, College of Medicine, 103 Hospital Dr., Saskatoon, SK, S7N 0W8, Canada
| | - Wayne Hill
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, College of Medicine, Saskatoon, SK, Canada
| | - Ayman El-Sayed
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, College of Medicine, Saskatoon, SK, Canada
| | - Kris Barreto
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, College of Medicine, Saskatoon, SK, Canada
| | - Clarence Ronald Geyer
- Department of Pathology and Laboratory Medicine, University of Saskatchewan, College of Medicine, Saskatoon, SK, Canada
| | - Humphrey Fonge
- Department of Medical Imaging, RUH Saskatoon, University of Saskatchewan, College of Medicine, 103 Hospital Dr., Saskatoon, SK, S7N 0W8, Canada. .,Department of Medical Imaging, Royal University Hospital Saskatoon, Saskatoon, SK, Canada.
| |
Collapse
|
23
|
Heregulin Drives Endocrine Resistance by Altering IL-8 Expression in ER-Positive Breast Cancer. Int J Mol Sci 2020; 21:ijms21207737. [PMID: 33086721 PMCID: PMC7589856 DOI: 10.3390/ijms21207737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/04/2023] Open
Abstract
Sustained HER2/HER3 signaling due to the overproduction of the HER3 ligand heregulin (HRG) is proposed as a key contributor to endocrine resistance in estrogen receptor-positive (ER+) breast cancer. The molecular mechanisms linking HER2 transactivation by HRG-bound HER3 to the acquisition of a hormone-independent phenotype in ER+ breast cancer is, however, largely unknown. Here, we explored the possibility that autocrine HRG signaling drives cytokine-related endocrine resistance in ER+ breast cancer cells. We used human cytokine antibody arrays to semi-quantitatively measure the expression level of 60 cytokines and growth factors in the extracellular milieu of MCF-7 cells engineered to overexpress full-length HRGβ2 (MCF-7/HRG cells). Interleukin-8 (IL-8), a chemokine closely linked to ER inaction, emerged as one the most differentially expressed cytokines. Cytokine profiling using structural deletion mutants lacking both the N-terminus and the cytoplasmic-transmembrane region of HRGβ2-which is not secreted and cannot transactivate HER2-or lacking a nuclear localization signal at the N-terminus-which cannot localize at the nucleus but is actively secreted and transactivates HER2-revealed that the HRG-driven activation of IL-8 expression in ER+ cells required HRG secretion and transactivation of HER2 but not HRG nuclear localization. The functional blockade of IL-8 with a specific antibody inversely regulated ERα-driven transcriptional activation in endocrine-sensitive MCF-7 cells and endocrine-resistant MCF-7/HRG cells. Overall, these findings suggest that IL-8 participates in the HRG-driven endocrine resistance program in ER+/HER2- breast cancer and might illuminate a potential clinical setting for IL8- or CXCR1/2-neutralizing antibodies.
Collapse
|
24
|
Masoodi TA, Shaik NA, Burhan S, Shafi G, Talluri VR. RNA-Seq reveals skipping of exon 3 in a breast cancer patient carrying G118D PIK3CA mutation. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
25
|
Cairns J, Ingle JN, Dudenkov TM, Kalari KR, Carlson EE, Na J, Buzdar AU, Robson ME, Ellis MJ, Goss PE, Shepherd LE, Goodnature B, Goetz MP, Weinshilboum RM, Li H, Bari MG, Wang L. Pharmacogenomics of aromatase inhibitors in postmenopausal breast cancer and additional mechanisms of anastrozole action. JCI Insight 2020; 5:137571. [PMID: 32701512 PMCID: PMC7455128 DOI: 10.1172/jci.insight.137571] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 07/15/2020] [Indexed: 01/09/2023] Open
Abstract
Aromatase inhibitors (AIs) reduce breast cancer recurrence and prolong survival, but up to 30% of patients exhibit recurrence. Using a genome-wide association study of patients entered on MA.27, a phase III randomized trial of anastrozole versus exemestane, we identified a single nucleotide polymorphism (SNP) in CUB And Sushi multiple domains 1 (CSMD1) associated with breast cancer–free interval, with the variant allele associated with fewer distant recurrences. Mechanistically, CSMD1 regulates CYP19 expression in an SNP- and drug-dependent fashion, and this regulation is different among 3 AIs: anastrozole, exemestane, and letrozole. Overexpression of CSMD1 sensitized AI-resistant cells to anastrozole but not to the other 2 AIs. The SNP in CSMD1 that was associated with increased CSMD1 and CYP19 expression levels increased anastrozole sensitivity, but not letrozole or exemestane sensitivity. Anastrozole degrades estrogen receptor α (ERα), especially in the presence of estradiol (E2). ER+ breast cancer organoids and AI- or fulvestrant-resistant breast cancer cells were more sensitive to anastrozole plus E2 than to AI alone. Our findings suggest that the CSMD1 SNP might help to predict AI response, and anastrozole plus E2 serves as a potential new therapeutic strategy for patients with AI- or fulvestrant-resistant breast cancers. A germline variation within the CSMD1 gene predicts aromatase inhibitor response in breast cancer.
Collapse
Affiliation(s)
- Junmei Cairns
- Department of Molecular Pharmacology and Experimental Therapeutics
| | | | - Tanda M Dudenkov
- Department of Molecular Pharmacology and Experimental Therapeutics
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin E Carlson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Jie Na
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, USA
| | - Aman U Buzdar
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mark E Robson
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Paul E Goss
- Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Barbara Goodnature
- Patient advocate, Mayo Clinic Breast Cancer Specialized Program of Research Excellence, Rochester, Minnesota, USA
| | | | | | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics
| | | | - Liewei Wang
- Department of Molecular Pharmacology and Experimental Therapeutics
| |
Collapse
|
26
|
Chang YC, Cheung CHA, Kuo YL. Tamoxifen Rechallenge Decreases Metastatic Potential but Increases Cell Viability and Clonogenicity in a Tamoxifen-Mediated Cytotoxicity-Resistant Subline of Human Breast MCF7 Cancer Cells. Front Cell Dev Biol 2020; 8:485. [PMID: 32695778 PMCID: PMC7338790 DOI: 10.3389/fcell.2020.00485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/22/2020] [Indexed: 12/17/2022] Open
Abstract
Background Drug resistance is frequently found in estrogen receptor-positive (ER+) breast cancer patients during and after prolonged tamoxifen treatment. Although tamoxifen rechallenge has been proposed for treating recurrent breast tumors, the clinical benefit of this treatment is still controversial. The aims of this study are to identify the possible tamoxifen cytotoxicity-resistant subpopulation of MCF7 cells and to determine the effects of tamoxifen rechallenge on these cells. Methods Western blot analysis was used to determine the expression levels of various epithelial-mesenchymal transition- and cell survival/proliferation-related proteins in MCF7 and MCF7-derived, tamoxifen-mediated cytotoxicity-resistant MCF7-TAM12.5 breast cancer cells. Wound healing, Transwell migration, and invasion assays were used to examine the metastatic potential of cells. Clonogenic assays, trypan blue exclusion assays, and bromodeoxyuridine assays were used to examine clonogenicity and to determine the proliferation rate of cells. Results We found that MCF7-TAM12.5 cells exhibited higher tolerance to tamoxifen-mediated cytotoxicity, higher metastatic potential, higher expression levels of XIAP, and lower expression levels of ERα/ERβ/HER2/Smac than MCF7 cells. In addition, MCF7 cells endogenously expressed Bcl-2α, whereas MCF7-TAM12.5 cells only expressed Bcl-2β. Interestingly, tamoxifen rechallenge decreased the metastatic potential but increased the proliferation and clonogenicity of MCF7-TAM12.5 cells. At the molecular level, tamoxifen rechallenge upregulated the expression of phosphorylated Aurora A and Aurora B kinase in MCF7-TAM12.5 cells. Conclusion Our findings further support the existence of highly heterogenetic cancer cell populations in ER+ breast tumors. It will be of clinical importance to determine the protein expression and the genetic profiles of tamoxifen-resistant/recurrent ER+ breast tumors to predict the potential effects of tamoxifen readministration in the future.
Collapse
Affiliation(s)
- Yung-Chieh Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chun Hei Antonio Cheung
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Lung Kuo
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan and Douliu, Taiwan
| |
Collapse
|
27
|
Huang B, Yip WK, Wei N, Luo KQ. Acetyltanshinone IIA is more potent than lapatinib in inhibiting cell growth and degrading HER2 protein in drug-resistant HER2-positive breast cancer cells. Cancer Lett 2020; 490:1-11. [PMID: 32585412 DOI: 10.1016/j.canlet.2020.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 10/24/2022]
Abstract
High expression of human epidermal factor receptor 2 (HER2) is directly related to tumor progression, malignancy and drug resistance in HER2-positive breast cancer (HER2-PBC). The major limitation of current anti-HER2 therapies is that they cannot reduce the levels of HER2 protein. Here, we investigated the effect of acetyltanshinone IIA (ATA) in lapatinib-resistant HER2-PBC cells. Our data showed that ATA exhibited more potent effects than lapatinib against drug-resistant HER2-PBC cells in terms of (1) inhibiting cell growth, (2) reducing phosphorylated and total HER2 levels, (3) inhibiting tumor xenograft growth in nude mice, and (4) reducing HER2 protein levels in tumor xenografts. A mechanistic study revealed that ATA promoted HER2 degradation via increasing c-Cbl and CHIP-mediated HER2 ubiquitination and subsequent HER2 degradation by the proteasome or lysosome. ATA also reduced the levels of other tyrosine kinase receptors (TKRs), such as HER3, IGF-1R and MET, in lapatinib-resistant cells. Our findings suggest that direct degradation of HER2 and other TKRs can be an effective strategy for combatting drug resistance. They also indicate the potential utilization of ATA in treating breast cancer that is resistant or nonresponsive to current HER2-targeted therapies.
Collapse
Affiliation(s)
- Bin Huang
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Wai Kien Yip
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China
| | - Na Wei
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR, China.
| |
Collapse
|
28
|
N083E (Alliance): long-term outcomes of patients treated in a pilot phase II study of docetaxel, carboplatin, trastuzumab, and lapatinib as adjuvant therapy for early-stage HER2-positive breast cancer. Breast Cancer Res Treat 2020; 182:613-622. [PMID: 32504284 DOI: 10.1007/s10549-020-05709-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/26/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND The addition of lapatinib (L) to trastuzumab (T) was previously found to be synergistic in preclinical models and in the neoadjuvant setting. Prior to the results of the ALTTO trial, this study assessed the safety and feasibility of adding L to the standard adjuvant docetaxel, carboplatin, and trastuzumab (TCH) regimen in early-stage HER2-positive breast cancer (HER2+ BC). METHODS In this single-arm, 2-stage, phase II study, patients with stages I-III HER2+ BC received TCH plus L at 1000 mg daily for a total of 12 months. The primary endpoint was the safety and tolerability, including the rate of diarrhea. Secondary endpoints included adverse event (AE) profile using the NCI CTCAE v3.0 and cardiac safety. RESULTS Thirty eligible patients were enrolled. Median follow-up is 5.3 years. Diarrhea was the most common AE with 50% Grade (G)1/2 and 43% G3 diarrhea. However, it was responsive to dose reduction of L (750 mg) and institution of anti-diarrheal medications. Cardiovascular AE were infrequent and no patients experienced congestive heart failure while on treatment. CONCLUSION TCHL was a tolerable regimen at a starting L dose of 750 mg PO daily when given concurrently with chemotherapy.
Collapse
|
29
|
Hanker AB, Sudhan DR, Arteaga CL. Overcoming Endocrine Resistance in Breast Cancer. Cancer Cell 2020; 37:496-513. [PMID: 32289273 PMCID: PMC7169993 DOI: 10.1016/j.ccell.2020.03.009] [Citation(s) in RCA: 445] [Impact Index Per Article: 111.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022]
Abstract
Estrogen receptor-positive (ER+) breast cancer is the most common breast cancer subtype. Treatment of ER+ breast cancer comprises interventions that suppress estrogen production and/or target the ER directly (overall labeled as endocrine therapy). While endocrine therapy has considerably reduced recurrence and mortality from breast cancer, de novo and acquired resistance to this treatment remains a major challenge. An increasing number of mechanisms of endocrine resistance have been reported, including somatic alterations, epigenetic changes, and changes in the tumor microenvironment. Here, we review recent advances in delineating mechanisms of resistance to endocrine therapies and potential strategies to overcome such resistance.
Collapse
Affiliation(s)
- Ariella B Hanker
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Dhivya R Sudhan
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Carlos L Arteaga
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
30
|
Meng B, Folaron MR, Byrd BK, Samkoe KS, Strawbridge RS, Barth C, Gibbs SL, Davis SC. Topical dual-probe staining using quantum dot-labeled antibodies for identifying tumor biomarkers in fresh specimens. PLoS One 2020; 15:e0230267. [PMID: 32160634 PMCID: PMC7065915 DOI: 10.1371/journal.pone.0230267] [Citation(s) in RCA: 3] [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: 10/11/2019] [Accepted: 02/25/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose Rapid, intra-operative identification of tumor tissue in the margins of excised specimens has become an important focus in the pursuit of reducing re-excision rates, especially for breast conserving surgery. Dual-probe difference specimen imaging (DDSI) is an emerging approach that uses the difference in uptake/clearance kinetics between a pair of fluorescently-labeled stains, one targeted to a biomarker-of-interest and the other an untargeted isotype, to reveal receptor-specific images of the specimen. Previous studies using antibodies labeled with either enhanced Raman particles or organic fluorophores have shown promising tumor vs. normal diagnostic performance. Yet, the unique properties of quantum dot-labeled antibody complexes (QDACs), which provide spectrally-distinct fluorescence emission from a common excitation source, make them ideal candidates for this application. Herein, we evaluate the diagnostic performance of QDAC-based DDSI in excised xenografts. Procedures Excised fresh specimens of normal tissue and human tumor xenografts with elevated expression of HER2 were stained with a HER2-targeted QDAC and an untargeted QDAC isotype. Stained specimens were imaged on a custom hyperspectral imaging system capable of spectrally separating the quantum dot signatures, and images processed using the DDSI approach. The diagnostic performance of this technique under different incubation temperatures and probe concentrations was evaluated using receiver-operator characteristic analysis. Results HER2-targeted QDAC-DDSI was able to distinguish HER2(+) tumors from normal tissue with reasonably high diagnostic performance; however, this performance was sensitive to temperature during the staining procedure. Area under the curve values were 0.61 when staining at room temperature but increased to over 0.81 when staining at 37 °C. Diagnostic performance was not affected by increasing stain concentration. Conclusions This study is the first to report dual-probe difference imaging of specimens using QDACs and hyperspectral imaging. Our results show promising diagnostic performance under certain conditions, and compel further optimization and evaluation of this intra-operative margin assessment technique.
Collapse
Affiliation(s)
- Boyu Meng
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Margaret R. Folaron
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Brook K. Byrd
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Kimberley S. Samkoe
- Geisel School of Medicine at Dartmouth College, Hanover, New Hampshire, United States of America
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
| | - Rendall S. Strawbridge
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States of America
| | - Connor Barth
- Biomedical Engineering Department, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Summer L. Gibbs
- Biomedical Engineering Department, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Scott C. Davis
- Thayer School of Engineering at Dartmouth College, Hanover, New Hampshire, United States of America
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, United States of America
- * E-mail:
| |
Collapse
|
31
|
Scales SJ, Tsai SP, Zacharias N, Cruz-Chuh JD, Bullen G, Velasquez E, Chang J, Bruguera E, Kozak KR, Sadowsky J. Development of a Cysteine-Conjugatable Disulfide FRET Probe: Influence of Charge on Linker Cleavage and Payload Trafficking for an Anti-HER2 Antibody Conjugate. Bioconjug Chem 2019; 30:3046-3056. [PMID: 31726009 DOI: 10.1021/acs.bioconjchem.9b00678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Disulfide-linked bioconjugates allow the delivery of pharmacologically active or other cargo to specific tissues in a redox-sensitive fashion. However, an understanding of the kinetics, subcellular distribution, and mechanism of disulfide cleavage in such bioconjugates is generally lacking. Here, we report a modular disulfide-linked TAMRA-BODIPY based FRET probe that can be readily synthesized, modified, and conjugated to a cysteine-containing biomolecule to enable real-time monitoring of disulfide cleavage during receptor-mediated endocytosis in cells. We demonstrate the utility of this probe to study disulfide reduction during HER2 receptor-mediated uptake of a Cys-engineered anti-HER2 THIOMAB antibody. We found that introduction of positive, but not negative, charges in the probe improved retention of the BODIPY catabolite. This permitted the observation of significant disulfide cleavage in endosomes or lysosomes on par with proteolytic cleavage of a similarly charged valine-citrulline peptide-based probe. In general, the FRET probe we describe should enable real-time cellular monitoring of disulfide cleavage in other targeted delivery systems for mechanistic or diagnostic applications. Furthermore, modifications to the released BODIPY moiety permit evaluation of physicochemical properties that govern lysosomal egress or retention, which may have implications for the development of next-generation antibody-drug conjugates.
Collapse
Affiliation(s)
- Suzie J Scales
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Siao Ping Tsai
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Neelie Zacharias
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Josefa Dela Cruz-Chuh
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Gordy Bullen
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Erick Velasquez
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Julie Chang
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Elise Bruguera
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Katherine R Kozak
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| | - Jack Sadowsky
- Research & Early Development , Genentech, Inc. , 1 DNA Way , South San Francisco , California 94080 , United States
| |
Collapse
|
32
|
Smyth LM, Piha-Paul SA, Won HH, Schram AM, Saura C, Loi S, Lu J, Shapiro GI, Juric D, Mayer IA, Arteaga CL, de la Fuente MI, Brufksy AM, Spanggaard I, Mau-Sørensen M, Arnedos M, Moreno V, Boni V, Sohn J, Schwartzberg LS, Gonzàlez-Farré X, Cervantes A, Bidard FC, Gorelick AN, Lanman RB, Nagy RJ, Ulaner GA, Chandarlapaty S, Jhaveri K, Gavrila EI, Zimel C, Selcuklu SD, Melcer M, Samoila A, Cai Y, Scaltriti M, Mann G, Xu F, Eli LD, Dujka M, Lalani AS, Bryce R, Baselga J, Taylor BS, Solit DB, Meric-Bernstam F, Hyman DM. Efficacy and Determinants of Response to HER Kinase Inhibition in HER2-Mutant Metastatic Breast Cancer. Cancer Discov 2019; 10:198-213. [PMID: 31806627 DOI: 10.1158/2159-8290.cd-19-0966] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 10/23/2019] [Accepted: 12/02/2019] [Indexed: 11/16/2022]
Abstract
HER2 mutations define a subset of metastatic breast cancers with a unique mechanism of oncogenic addiction to HER2 signaling. We explored activity of the irreversible pan-HER kinase inhibitor neratinib, alone or with fulvestrant, in 81 patients with HER2-mutant metastatic breast cancer. Overall response rate was similar with or without estrogen receptor (ER) blockade. By comparison, progression-free survival and duration of response appeared longer in ER+ patients receiving combination therapy, although the study was not designed for direct comparison. Preexistent concurrent activating HER2 or HER3 alterations were associated with poor treatment outcome. Similarly, acquisition of multiple HER2-activating events, as well as gatekeeper alterations, were observed at disease progression in a high proportion of patients deriving clinical benefit from neratinib. Collectively, these data define HER2 mutations as a therapeutic target in breast cancer and suggest that coexistence of additional HER signaling alterations may promote both de novo and acquired resistance to neratinib. SIGNIFICANCE: HER2 mutations define a targetable breast cancer subset, although sensitivity to irreversible HER kinase inhibition appears to be modified by the presence of concurrent activating genomic events in the pathway. These findings have implications for potential future combinatorial approaches and broader therapeutic development for this genomically defined subset of breast cancer.This article is highlighted in the In This Issue feature, p. 161.
Collapse
Affiliation(s)
- Lillian M Smyth
- Memorial Sloan Kettering Cancer Center, New York, New York.,St. Vincent's University Hospital, Dublin, Ireland
| | | | - Helen H Won
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Cristina Saura
- Vall d'Hebron University Hospital, Vall d'Hebrón Institute of Oncology (VHIO), Barcelona, Spain
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Janice Lu
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, California
| | | | - Dejan Juric
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Carlos L Arteaga
- The University of Texas Southwestern Medical Center Harold C. Simmons Comprehensive Cancer Center, Dallas, Texas
| | | | - Adam M Brufksy
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | | | | | | | - Valentina Boni
- START Madrid Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Joohyuk Sohn
- Yonsei Cancer Center, University College of Medicine, Seoul, Korea
| | | | | | - Andrés Cervantes
- CIBERONC, Biomedical Research Institute INCLIVA, University of Valencia, Valencia, Spain
| | | | | | | | | | - Gary A Ulaner
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | | | | | - Myra Melcer
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Yanyan Cai
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Grace Mann
- Puma Biotechnology, Inc., Los Angeles, California
| | - Feng Xu
- Puma Biotechnology, Inc., Los Angeles, California
| | - Lisa D Eli
- Puma Biotechnology, Inc., Los Angeles, California
| | | | | | | | - José Baselga
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Barry S Taylor
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - David B Solit
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - David M Hyman
- Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
33
|
Rajaratinam H, Nafi SNM. Andrographolide is an Alternative Treatment to Overcome Resistance in ER-Positive Breast Cancer via Cholesterol Biosynthesis Pathway. Malays J Med Sci 2019; 26:6-20. [PMID: 31728115 PMCID: PMC6839656 DOI: 10.21315/mjms2019.26.5.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 03/17/2019] [Indexed: 12/13/2022] Open
Abstract
Oestrogen receptor (ER)-positive breast cancer is one of the common forms of breast cancer affecting women worldwide. ER-positive breast cancer patients are subjected to anti-oestrogen therapy such as selective oestrogen receptor modulator (SERM) and aromatase inhibitors (AIs). Recently, the emergence of resistance to anti-oestrogen treatment is under intensive focus. The different mechanisms postulated to explain the occurrence of resistance in ER-positive breast cancer treatment include the loss of ER function and the crosstalk between signalling pathways in cancer cells. Recent literature highlighted that the cholesterol biosynthesis pathway acts as a novel mechanism underlying resistance to oestrogen deprivation. The present study aimed to highlight the role of cholesterol biosynthesis in anti-oestrogen treatment resistance, putatively suggesting an alternative plant-based treatment using andrographolide from Andrographis paniculata. The hypolipidaemic effect of andrographolide can be utilised to prevent the resistance in the treatment of ER-positive breast cancer contributed by cholesterol biosynthesis.
Collapse
Affiliation(s)
- Harishini Rajaratinam
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - Siti Norasikin Mohd Nafi
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Pathology, Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
34
|
Parmar V, Nair NS, Thakkar P, Chitkara G. Molecular Biology in the Breast Clinics-Current status and future perspectives. Indian J Surg Oncol 2019; 12:7-20. [PMID: 33994723 DOI: 10.1007/s13193-019-00954-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 07/19/2019] [Indexed: 10/26/2022] Open
Abstract
Breast cancer is no longer considered a single disease, and with better understanding of cancer biology, its management has evolved over the years, into a complex individualized use of therapeutics based on variable expressions of predictive and prognostic factors. With the advent of molecular and genetic research, the complexity and diversity of breast cancer cells and their ability to survive and develop resistance to treatment strategies became more evident. At the same time, targeted therapies evolved, as specific targets were discovered such as HER2 receptor, and androgen receptor. More recent is the development of immunotherapy which aims at strengthening the host immune system to identify and kill the tumor cells. In breast cancer treatment, use of molecular tests has been a target of controversies, due to their high costs and inaccessibility in limited resource situations. Research in breast cancer is also proceeding at a rapid pace, but it is important to remember that breast cancer continues to be a complex interplay of alterations at molecular and genetic level, with the variability in expressions at protein level leading to difference in behavior and responses to treatment and overall outcome. In the succeeding paragraphs, we will try to review the available evidence in literature and attempt to understand the molecular complexity of breast cancer in order to simplify the art of treating the disease and improving outcomes.
Collapse
Affiliation(s)
- Vani Parmar
- Breast Unit, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra 410210 India
| | - Nita S Nair
- Breast Unit, Tata Memorial Centre, Tata Memorial Hospital, Ernest Borges Rd, Parel, Mumbai, 400012 India
| | - Purvi Thakkar
- Breast Unit, Tata Memorial Centre, Tata Memorial Hospital, Ernest Borges Rd, Parel, Mumbai, 400012 India
| | - Garvit Chitkara
- Breast Unit, Tata Memorial Centre, Tata Memorial Hospital, Ernest Borges Rd, Parel, Mumbai, 400012 India
| |
Collapse
|
35
|
El Sayed R, El Jamal L, El Iskandarani S, Kort J, Abdel Salam M, Assi H. Endocrine and Targeted Therapy for Hormone-Receptor-Positive, HER2-Negative Advanced Breast Cancer: Insights to Sequencing Treatment and Overcoming Resistance Based on Clinical Trials. Front Oncol 2019; 9:510. [PMID: 31281796 PMCID: PMC6597942 DOI: 10.3389/fonc.2019.00510] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/28/2019] [Indexed: 01/30/2023] Open
Abstract
Background: Advanced hormone-receptor positive HER2 negative breast cancer is a common and a very heterogeneous disease. Hormone therapy is the main first line treatment of choice, given alone or in combination with other agents that have shown to improve patient outcomes, Nevertheless, treatment remains generally palliative rather than curative. Sequencing of such treatment remains challenging, especially with resurgence of variable resistance patterns. Multiple attempts have been made to overcome resistance and improve patient survival, yet resistance remains not very well understood and metastatic cancer remains a disease with dismal prognosis. Methods: In this paper, we searched pubmed database as well as local and international meetings for all studies discussing advanced and metastatic hormone-receptor-positive, her2-negative breast cancer, hormonal treatment, resistance to hormonal treatment, mechanism of resistance, and means to overcome such resistance. Conclusion: There does not exist an optimal treatment sequence for hormone-receptor-positive, her2-negative advanced breast cancer. However, after review of literature, a reasonable approach may be starting with tamoxifen, aromatase inhibitors, or fulvestrant in absence of visceral crisis, in addition to ensuring adequate ovarian function suppression in pre/peri-menopausal women. Aromatase inhibitors and fulvestrant seem to be superior. Resistance to such agents is increasing, mostly attributed to genetic and molecular changes. Multiple modalities are addressed to overcome such resistance including use of CKD4/6 inhibitors, mTOR inhibitors and PI3K inhibitors in addition to other agents under study, all with promising results. CDK4/6 inhibitors work best when used in frontline setting. Finally, treatment of breast cancer remains a growing field, and more studies are to be awaited.
Collapse
Affiliation(s)
- Rola El Sayed
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| | - Lara El Jamal
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Jeries Kort
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| | | | - Hazem Assi
- Oncology Division, Department of Internal Medicine, American University of Beirut Medical Center, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
36
|
El-Abd E, Matta CA, Sheta M, El-Kerm Y, Sakr S. Histopathological characteristics of breast cancer and evaluation of ER alpha and Her-2neu using immunohistochemical and RT-PCR techniques. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2014.03.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Eman El-Abd
- Molecular Biology Department, Medical Technology Centre (MTC), Medical Research Institute (MRI), Alexandria University, Egypt
- Radiation Sciences Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Cecil A. Matta
- Zoology Department, Faculty of Science, Alexandria University, Egypt
| | - Manal Sheta
- Pathology Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Yasser El-Kerm
- Cancer Research and Management Department, Medical Research Institute (MRI), Alexandria University, Egypt
| | - Shimaa Sakr
- Medical Technology Centre (MTC), Medical Research Institute (MRI), Alexandria University, Egypt
| |
Collapse
|
37
|
Cui X, Chen H, Zhang Q, Xu M, Yuan G, Zhou J. Exploration of the Structure and Recognition of a G-quadruplex in the her2 Proto-oncogene Promoter and Its Transcriptional Regulation. Sci Rep 2019; 9:3966. [PMID: 30850693 PMCID: PMC6408435 DOI: 10.1038/s41598-019-39941-5] [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: 10/05/2018] [Accepted: 01/23/2019] [Indexed: 12/02/2022] Open
Abstract
G-quadruplexes in oncogene promoters provide putative targets for transcriptional regulation. The structure of a putative G-quadruplex sequence (S1: GGAGAAGGAGGAGGTGGAGGAGGAGGG) in potassium solution in the her2 promoter has been resolved mainly through nuclear magnetic resonance (NMR) spectroscopy. By application of various NMR spectra, we proved the formation of a four-layer G-quadruplex composing of two G-tetrads and two G/A-mixed planes with a four-residues loop (A3-G4-A5-A6). Further evidence from a luciferase reporter assay, Q-RT-PCR and Western blotting indicates that S1 G-quadruplex formation can repress her2 promoter activity, and a selected G-quadruplex ligand cβ can enhance the repression by down regulating her2 transcription and expression. These findings provide a G-quadruplex target and perspective implications in her2 transcriptional regulation.
Collapse
Affiliation(s)
- Xiaojie Cui
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China. .,College of Life and Environmental Sciences, Minzu University of China, Beijing, 100081, China.
| | - Han Chen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Qiang Zhang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Department of Cardiology, Peking University Third Hospital, Beijing, 100191, China
| | - Gu Yuan
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China
| | - Jiang Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
38
|
Salmerón-Hernández Á, Noriega-Reyes MY, Jordan A, Baranda-Avila N, Langley E. BCAS2 Enhances Carcinogenic Effects of Estrogen Receptor Alpha in Breast Cancer Cells. Int J Mol Sci 2019; 20:ijms20040966. [PMID: 30813351 PMCID: PMC6412365 DOI: 10.3390/ijms20040966] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/11/2019] [Accepted: 02/21/2019] [Indexed: 01/18/2023] Open
Abstract
Estrogen receptor alpha (ERα) has an established role in breast cancer biology. Transcriptional activation by ERα is a multistep process modulated by coactivator and corepressor proteins. Breast Cancer Amplified Sequence 2 (BCAS2), is a poorly studied ERα coactivator. In this work, we characterize some of the mechanisms through which this protein increases ERα activity and how this promotes carcinogenic processes in breast cancer cells. Using protein-protein interaction and luciferase assays we show that BCAS2 interacts with ERα both in vitro and in vivo and upregulates transcriptional activation of ERα directly through its N-terminal region (AF-1) and indirectly through its C-terminal (AF-2) region, acting in concert with AF-2 interacting coactivators. Elevated expression of BCAS2 positively affects proliferation, clonogenicity and migration of breast cancer cells and directly activates ERα regulated genes which have been shown to play a role in tumor growth and progression. Finally, we used signal transduction pathway inhibitors to elucidate how BCAS2 is regulated in these cells and observed that BCAS2 is preferentially regulated by the PI3K/AKT signaling pathway. BCAS2 is an AF-1 coactivator of ERα whose overexpression promotes carcinogenic processes, suggesting an important role in the development of estrogen-receptor positive breast cancer.
Collapse
Affiliation(s)
- Ángel Salmerón-Hernández
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, 14080 Mexico City, Mexico.
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| | - María Yamilet Noriega-Reyes
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, 14080 Mexico City, Mexico.
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, 04510 Mexico City, Mexico.
| | - Albert Jordan
- Institut de Biología Molecular de Barcelona (IBMB-CSIC) Parc Científic de Barcelona, Barcelona, 08028 Cataluña, Spain.
| | - Noemi Baranda-Avila
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, 14080 Mexico City, Mexico.
| | - Elizabeth Langley
- Departamento de Investigación Básica, Instituto Nacional de Cancerología, Av. San Fernando No. 22, Col. Sección XVI, 14080 Mexico City, Mexico.
| |
Collapse
|
39
|
Jarman EJ, Ward C, Turnbull AK, Martinez-Perez C, Meehan J, Xintaropoulou C, Sims AH, Langdon SP. HER2 regulates HIF-2α and drives an increased hypoxic response in breast cancer. Breast Cancer Res 2019; 21:10. [PMID: 30670058 PMCID: PMC6343358 DOI: 10.1186/s13058-019-1097-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Accepted: 01/04/2019] [Indexed: 12/21/2022] Open
Abstract
Background Tumour hypoxia is a driver of breast cancer progression associated with worse prognosis and more aggressive disease. The cellular response to hypoxia is mediated by the hypoxia-inducible transcription factors HIF-1 and HIF-2, whose transcriptional activity is canonically regulated through their oxygen-labile HIF-α subunits. These are constitutively degraded in the presence of oxygen; however, HIF-1α can be stabilised, even at high oxygen concentrations, through the activation of HER receptor signalling. Despite this, there is still limited understanding on how HER receptor signalling interacts with HIF activity to contribute to breast cancer progression in the context of tumour hypoxia. Methods 2D and 3D cell line models were used alongside microarray gene expression analysis and meta-analysis of publicly available gene expression datasets to assess the impact of HER2 overexpression on HIF-1α/HIF-2α regulation and to compare the global transcriptomic response to acute and chronic hypoxia in an isogenic cell line model of HER2 overexpression. Results HER2 overexpression in MCF7 cells leads to an increase in HIF-2α but not HIF-1α expression in normoxia and an increased upregulation of HIF-2α in hypoxia. Global gene expression analysis showed that HER2 overexpression in these cells promotes an exaggerated transcriptional response to both short-term and long-term hypoxia, with increased expression of numerous hypoxia response genes. HIF-2α expression is frequently higher in HER2-overexpressing tumours and is associated with worse disease-specific survival in HER2-positive breast cancer patients. HER2-overexpressing cell lines demonstrate an increased sensitivity to targeted HIF-2α inhibition through either siRNA or the use of a small molecule inhibitor of HIF-2α translation. Conclusions This study suggests an important interplay between HER2 expression and HIF-2α in breast cancer and highlights the potential for HER2 to drive the expression of numerous hypoxia response genes in normoxia and hypoxia. Overall, these findings show the importance of understanding the regulation of HIF activity in a variety of breast cancer subtypes and points to the potential of targeting HIF-2α as a therapy for HER2-positive breast cancer. Electronic supplementary material The online version of this article (10.1186/s13058-019-1097-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edward J Jarman
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK. .,Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road South, Edinburgh, EH4 2XU, UK.
| | - Carol Ward
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Arran K Turnbull
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Carlos Martinez-Perez
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Chrysi Xintaropoulou
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, EH4 2XR, UK
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| |
Collapse
|
40
|
Haque MM, Desai KV. Pathways to Endocrine Therapy Resistance in Breast Cancer. Front Endocrinol (Lausanne) 2019; 10:573. [PMID: 31496995 PMCID: PMC6712962 DOI: 10.3389/fendo.2019.00573] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/06/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancers with positive expression of Estrogen Receptor (ER+) are treated with anti-hormone/endocrine therapy which targets the activity of the receptor, the half-life of the receptor or the availability of estrogen. This has significantly decreased mortality in women with ER+ breast cancer, however, about 25-30% of treated women run the risk or recurrence due to either intrinsic or acquired resistance to endocrine therapies. While ER itself is a predictor of response to such therapies, there exists a need to find more biomarkers and novel targets to treat resistant tumors. In this review, we summarize the known mechanisms and describe the ability of genomics in unraveling rare mutations and gene rearrangements that may impact the development of resistance and therefore treatment of ER+ breast cancer in the near future.
Collapse
|
41
|
Sen F, Aydiner A. Endocrine Therapy of Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Tyagi A, Liu X, Abidi IH, Gao Z, Park BM, Zeng X, Ou X, Cagang AA, Zhuang M, Hossain MD, Zhang K, Weng LT, Sun F, Luo Z. Modular functionalization of crystalline graphene by recombinant proteins: a nanoplatform for probing biomolecules. NANOSCALE 2018; 10:22572-22582. [PMID: 30480695 DOI: 10.1039/c8nr06225k] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Graphene, as well as other two-dimensional materials, is a promising candidate for use in bioimaging, therapeutic drug delivery, and bio-sensing applications. Here, we developed a protocol to functionalize graphene with recombinant proteins using genetically encoded SpyTag-SpyCatcher chemistry. SpyTag forms a covalent isopeptide bond with its genetically encoded partner SpyCatcher through spontaneous amidation under physiological conditions. The functionalization protocol developed is based on the use of short proteins as a linker, where two graphene-binding-peptides (GBPs) are attached to both ends of SpyTag (referred to as GStG), followed by the covalent conjugation with SpyCatcher-fusion proteins. The proposed method enables the decoration of crystalline graphene with various proteins, such as fluorescent proteins and affibody molecules that bind to cancerous cells. This scheme, which takes advantage of the cleanness of single-crystal graphene and the robustness of SpyTag-SpyCatcher chemistry, provides a versatile platform on which to study the biomolecule-surface and cell-substrate interactions and, indeed, may lead to a new way of designing biomedical devices. The interaction between peptides and graphene was clearly shown using molecular dynamics simulation and proven using specially designed experiments.
Collapse
Affiliation(s)
- Abhishek Tyagi
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Nayar U, Cohen O, Kapstad C, Cuoco MS, Waks AG, Wander SA, Painter C, Freeman S, Persky NS, Marini L, Helvie K, Oliver N, Rozenblatt-Rosen O, Ma CX, Regev A, Winer EP, Lin NU, Wagle N. Acquired HER2 mutations in ER + metastatic breast cancer confer resistance to estrogen receptor-directed therapies. Nat Genet 2018; 51:207-216. [PMID: 30531871 DOI: 10.1038/s41588-018-0287-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Seventy percent of breast cancers express the estrogen receptor (ER), and agents that target the ER are the mainstay of treatment. However, virtually all people with ER+ breast cancer develop resistance to ER-directed agents in the metastatic setting. Beyond mutations in the ER itself, which occur in 25-30% of people treated with aromatase inhibitors1-4, knowledge about clinical resistance mechanisms remains incomplete. We identified activating HER2 mutations in metastatic biopsies from eight patients with ER+ metastatic breast cancer who had developed resistance to aromatase inhibitors, tamoxifen or fulvestrant. Examination of treatment-naive primary tumors in five patients showed no evidence of pre-existing mutations in four of five patients, suggesting that these mutations were acquired under the selective pressure of ER-directed therapy. The HER2 mutations and ER mutations were mutually exclusive, suggesting a distinct mechanism of acquired resistance to ER-directed therapies. In vitro analysis confirmed that the HER2 mutations conferred estrogen independence as well as-in contrast to ER mutations-resistance to tamoxifen, fulvestrant and the CDK4 and CDK6 inhibitor palbociclib. Resistance was overcome by combining ER-directed therapy with the irreversible HER2 kinase inhibitor neratinib.
Collapse
Affiliation(s)
- Utthara Nayar
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ofir Cohen
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian Kapstad
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael S Cuoco
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adrienne G Waks
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Seth A Wander
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Samuel Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Lori Marini
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Karla Helvie
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Nelly Oliver
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Cynthia X Ma
- Division of Oncology, Department of Medicine, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Howard Hughes Medical Institute and Koch Institute of Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric P Winer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Nikhil Wagle
- Center for Cancer Precision Medicine, Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
44
|
You SH, Chae BJ, Eom YH, Yoo TK, Kim YS, Kim JS, Park WC. Clinical Differences in Triple-Positive Operable Breast Cancer Subtypes in Korean Patients: An Analysis of Korean Breast Cancer Registry Data. J Breast Cancer 2018; 21:415-424. [PMID: 30607163 PMCID: PMC6310716 DOI: 10.4048/jbc.2018.21.e53] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 11/01/2018] [Indexed: 12/15/2022] Open
Abstract
Purpose Triple-positive breast cancer is defined by estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2) positivity. Several systemic breast cancer therapies target hormonal and HER2 responsiveness. We compared clinical outcomes of triple-positive disease with those of HER2-enriched and luminal HER2-negative disease and investigated the clinical efficacy of anti-HER2 therapy for triple-positive disease. Methods We retrospectively compared overall and recurrence-free survival among cases included in the Korean Breast Cancer Society (KBCS) and Seoul St. Mary's Hospital breast cancer registries and the therapeutic efficacy of trastuzumab for triple-positive and HER2-enriched cases. Results KBCS registry data (2006–2010; median follow-up, 76 months) indicated that patients with triple-positive breast cancer had intermediate survival between those with luminal A and HER2-enriched subtypes (p<0.001). Trastuzumab did not improve overall survival among patients with triple-positive breast cancer (p=0.899) in contrast to the HER2-enriched subtype (p=0.018). Seoul St. Mary's Hospital registry data indicated similar recurrence-free survival outcomes (p<0.001) and a lack of improvement with trastuzumab among patients with triple-positive breast cancer (median follow-up, 33 months; p=0.800). Multivariate analysis revealed that patients with triple-positive breast cancer had better overall survival than those with HER2-enriched disease and similar survival as those with the luminal A subtype (triple-positive: hazard ratio, 1.258, p=0.118; HER2-enriched: hazard ratio, 2.377, p<0.001). Conclusion Our findings showed that anti-HER2 therapy was less beneficial for treatment of triple-positive breast cancer than for HER2-enriched subtypes of breast cancer, and the triple-positive subtype had a distinct prognosis.
Collapse
Affiliation(s)
- Sun Hyong You
- Division of Breast and Thyroid Surgery, Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Byung Joo Chae
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong Hwa Eom
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae-Kyung Yoo
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Seok Kim
- Division of Breast and Thyroid Surgery, Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Jeong Soo Kim
- Division of Breast and Thyroid Surgery, Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
45
|
Croessmann S, Formisano L, Kinch LN, Gonzalez-Ericsson PI, Sudhan DR, Nagy RJ, Mathew A, Bernicker EH, Cristofanilli M, He J, Cutler RE, Lalani AS, Miller VA, Lanman RB, Grishin NV, Arteaga CL. Combined Blockade of Activating ERBB2 Mutations and ER Results in Synthetic Lethality of ER+/HER2 Mutant Breast Cancer. Clin Cancer Res 2018; 25:277-289. [PMID: 30314968 DOI: 10.1158/1078-0432.ccr-18-1544] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/05/2018] [Accepted: 10/09/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE We examined the role of ERBB2-activating mutations in endocrine therapy resistance in estrogen receptor positive (ER+) breast cancer. EXPERIMENTAL DESIGN ERBB2 mutation frequency was determined from large genomic databases. Isogenic knock-in ERBB2 mutations in ER+ MCF7 cells and xenografts were used to investigate estrogen-independent growth. Structural analysis was used to determine the molecular interaction of HER L755S with HER3. Small molecules and siRNAs were used to inhibit PI3Kα, TORC1, and HER3. RESULTS Genomic data revealed a higher rate of ERBB2 mutations in metastatic versus primary ER+ tumors. MCF7 cells with isogenically incorporated ERBB2 kinase domain mutations exhibited resistance to estrogen deprivation and to fulvestrant both in vitro and in vivo, despite maintaining inhibition of ERα transcriptional activity. Addition of the irreversible HER2 tyrosine kinase inhibitor neratinib restored sensitivity to fulvestrant. HER2-mutant MCF7 cells expressed higher levels of p-HER3, p-AKT, and p-S6 than cells with wild-type HER2. Structural analysis of the HER2 L755S variant implicated a more flexible active state, potentially allowing for enhanced dimerization with HER3. Treatment with a PI3Kα inhibitor, a TORC1 inhibitor or HER3 siRNA, but not a MEK inhibitor, restored sensitivity to fulvestrant and to estrogen deprivation. Inhibition of mutant HER2 or TORC1, when combined with fulvestrant, equipotently inhibited growth of MCF7/ERBB2 V777L xenografts, suggesting a role for TORC1 in antiestrogen resistance induced by ERBB2 mutations. CONCLUSIONS ERBB2 mutations hyperactivate the HER3/PI3K/AKT/mTOR axis, leading to antiestrogen resistance in ER+ breast cancer. Dual blockade of the HER2 and ER pathways is required for the treatment of ER+/HER2 mutant breast cancers.
Collapse
Affiliation(s)
- Sarah Croessmann
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Luigi Formisano
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa N Kinch
- Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, Texas
| | - Paula I Gonzalez-Ericsson
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dhivya R Sudhan
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Aju Mathew
- University of Kentucky Markey Cancer Center, Lexington, Kentucky
| | | | | | - Jie He
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | | | | | | | - Nick V Grishin
- Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, Texas.,Department of Biophysics, UT Southwestern Medical Center, Dallas, Texas.,Department of Biochemistry, UT Southwestern Medical Center, Dallas, Texas
| | - Carlos L Arteaga
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee. .,Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.,Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.,Harold C. Simmons Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
46
|
Epigenetic Silencing of THY1 Tracks the Acquisition of the Notch1–EGFR Signaling in a Xenograft Model of CD44+/CD24low/CD90+ Myoepithelial Cells. Mol Cancer Res 2018; 17:628-641. [DOI: 10.1158/1541-7786.mcr-17-0324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/13/2017] [Accepted: 09/13/2018] [Indexed: 11/16/2022]
|
47
|
Shen Y, Fujii T, Ueno NT, Tripathy D, Fu N, Zhou H, Ning J, Xiao L. Comparative efficacy of adjuvant trastuzumab-containing chemotherapies for patients with early HER2-positive primary breast cancer: a network meta-analysis. Breast Cancer Res Treat 2018; 173:1-9. [PMID: 30242579 DOI: 10.1007/s10549-018-4969-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/15/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Trastuzumab (H) with chemotherapy benefits patients with HER2+ breast cancer (BC); however, we lack head-to-head pairwise assessment of survival or cardiotoxicity for specific combinations. We sought to identify optimal combinations. METHODS We searched PubMed, updated October 2017, using keywords "Breast Neoplasms/drug therapy," "Trastuzumab," and "Clinical Trial" and searched Cochrane Library. Our search included randomized trials of adjuvant H plus chemotherapy for early-stage HER2+ BC, and excluding trials of neoadjuvant therapy or without data to obtain hazard ratios (HRs) for outcomes. Following PRISMA guidelines, one investigator did initial search; two others independently confirmed and extracted information; and consensus with another investigator resolved disagreements. Before gathering data, we set outcomes of overall survival (OS), event-free survival (EFS), and severe cardiac adverse events (SCAEs). Analyzing 6 trials and 13,621 patients, we made direct and indirect comparisons using network meta-analysis on HR for OS or EFS and on odds ratio (OR) for SCAE; ranked therapy was done based on outcomes using p scores. RESULTS Compared with anthracycline-cyclophosphamide with taxane (ACT), ACT with concurrent H (ACT+H) showed best OS (HR 0.63, 95% confidence interval [CI] 0.55, 0.72), followed by taxane and carboplatin (TC) with concurrent H (TC+H) (HR 0.77, 95% CI 0.59, 1) and ACT with sequential H (ACT-H) (HR 0.85, 95% CI 0.68, 1.05). Pairwise comparisons showed statistically significant OS benefit for ACT+H over others; similar results for EFS. TC+H showed statistically significant lower SCAE risk compared to ACT+H (OR 0.13, 95% CI 0.03, 0.61). CONCLUSIONS Concurrent H with ACT or TC showed most clinical benefit for early-stage HER2+ BC; TC+H had lowest cardiotoxicity.
Collapse
Affiliation(s)
- Y Shen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1411, Houston, TX, 77030, USA.
| | - T Fujii
- Section of Translational Breast Cancer, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - N T Ueno
- Section of Translational Breast Cancer, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - D Tripathy
- Section of Translational Breast Cancer, Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - N Fu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1411, Houston, TX, 77030, USA
| | - H Zhou
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1411, Houston, TX, 77030, USA
| | - J Ning
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1411, Houston, TX, 77030, USA
| | - L Xiao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler St, Unit 1411, Houston, TX, 77030, USA
| |
Collapse
|
48
|
Schedin TB, Borges VF, Shagisultanova E. Overcoming Therapeutic Resistance of Triple Positive Breast Cancer with CDK4/6 Inhibition. Int J Breast Cancer 2018; 2018:7835095. [PMID: 30018827 PMCID: PMC6029445 DOI: 10.1155/2018/7835095] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/20/2018] [Accepted: 05/08/2018] [Indexed: 01/30/2023] Open
Abstract
Triple positive breast cancers overexpress both the human epidermal growth factor receptor 2 (HER2) oncogene and the hormonal receptors (HR) to estrogen and progesterone. These cancers represent a unique therapeutic challenge because of a bidirectional cross-talk between the estrogen receptor alpha (ERα) and HER2 pathways leading to tumor progression and resistance to targeted therapy. Attempts to combine standard of care HER2-targeted drugs with antihormonal agents for the treatment of HR+/HER2+ breast cancer yielded encouraging results in preclinical experiments but did improve overall survival in clinical trial. In this review, we dissect multiple mechanisms of therapeutic resistance typical of HR+/HER2+ breast cancer, summarize prior clinical trials of targeted agents, and describe novel rational drug combinations that include antihormonal agents, HER2-targeted drugs, and CDK4/6 inhibitors for treatment of the HR+/HER2+ breast cancer subtype.
Collapse
Affiliation(s)
- Troy B. Schedin
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Virginia F. Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| | - Elena Shagisultanova
- Young Women's Breast Cancer Translational Program, University of Colorado Denver, 13001 E 17th Pl, Aurora, CO 80045, USA
| |
Collapse
|
49
|
Fanning SW, Hodges-Gallagher L, Myles DC, Sun R, Fowler CE, Plant IN, Green BD, Harmon CL, Greene GL, Kushner PJ. Specific stereochemistry of OP-1074 disrupts estrogen receptor alpha helix 12 and confers pure antiestrogenic activity. Nat Commun 2018; 9:2368. [PMID: 29915250 PMCID: PMC6006285 DOI: 10.1038/s41467-018-04413-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 04/27/2018] [Indexed: 11/09/2022] Open
Abstract
Complex tissue-specific and cell-specific signaling by the estrogen receptor (ER) frequently leads to the development of resistance to endocrine therapy for breast cancer. Pure ER antagonists, which completely lack tissue-specific agonist activity, hold promise for preventing and treating endocrine resistance, however an absence of structural information hinders the development of novel candidates. Here we synthesize a small panel of benzopyrans with variable side chains to identify pure antiestrogens in a uterotrophic assay. We identify OP-1074 as a pure antiestrogen and a selective ER degrader (PA-SERD) that is efficacious in shrinking tumors in a tamoxifen-resistant xenograft model. Biochemical and crystal structure analyses reveal a structure activity relationship implicating the importance of a stereospecific methyl on the pyrrolidine side chain of OP-1074, particularly on helix 12. Estrogen receptor alpha (ERα) plays critical roles in the etiology and treatment of breast cancer. Here the authors synthesize benzopyrans with variable side chains to identify antiestrogenic compounds and characterize OP-1074, a compound that exhibits pure antiestrogenic activity by inducing the degradation of ERα and possesses greater potency than tamoxifen or fulvestrant in a xenograft model.
Collapse
Affiliation(s)
- S W Fanning
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA.
| | | | - D C Myles
- Olema Pharmaceuticals, San Francisco, CA, 94107, USA
| | - R Sun
- Olema Pharmaceuticals, San Francisco, CA, 94107, USA
| | - C E Fowler
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - I N Plant
- Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - B D Green
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - C L Harmon
- Olema Pharmaceuticals, San Francisco, CA, 94107, USA
| | - G L Greene
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL, 60637, USA
| | - P J Kushner
- Olema Pharmaceuticals, San Francisco, CA, 94107, USA
| |
Collapse
|
50
|
Boonyaratanakornkit V, Hamilton N, Márquez-Garbán DC, Pateetin P, McGowan EM, Pietras RJ. Extranuclear signaling by sex steroid receptors and clinical implications in breast cancer. Mol Cell Endocrinol 2018; 466:51-72. [PMID: 29146555 PMCID: PMC5878997 DOI: 10.1016/j.mce.2017.11.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022]
Abstract
Estrogen and progesterone play essential roles in the development and progression of breast cancer. Over 70% of breast cancers express estrogen receptors (ER) and progesterone receptors (PR), emphasizing the need for better understanding of ER and PR signaling. ER and PR are traditionally viewed as transcription factors that directly bind DNA to regulate gene networks. In addition to nuclear signaling, ER and PR mediate hormone-induced, rapid extranuclear signaling at the cell membrane or in the cytoplasm which triggers downstream signaling to regulate rapid or extended cellular responses. Specialized membrane and cytoplasmic proteins may also initiate hormone-induced extranuclear signaling. Rapid extranuclear signaling converges with its nuclear counterpart to amplify ER/PR transcription and specify gene regulatory networks. This review summarizes current understanding and updates on ER and PR extranuclear signaling. Further investigation of ER/PR extranuclear signaling may lead to development of novel targeted therapeutics for breast cancer management.
Collapse
Affiliation(s)
- Viroj Boonyaratanakornkit
- Department of Clinical Chemistry Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Age-related Inflammation and Degeneration Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Nalo Hamilton
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Diana C Márquez-Garbán
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Prangwan Pateetin
- Graduate Program in Clinical Biochemistry and Molecular Medicine, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Eileen M McGowan
- Chronic Disease Solutions Team, School of Life Sciences, University of Technology Sydney, Ultimo, 2007, Sydney, Australia
| | - Richard J Pietras
- UCLA Jonsson Comprehensive Cancer Center, Department of Medicine, Division of Hematology-Oncology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|