1
|
Daimon T, Bhattacharya A, Wang K, Haratake N, Nakashoji A, Ozawa H, Morimoto Y, Yamashita N, Kosaka T, Oya M, Kufe DW. MUC1-C is a target of salinomycin in inducing ferroptosis of cancer stem cells. Cell Death Discov 2024; 10:9. [PMID: 38182558 PMCID: PMC10770371 DOI: 10.1038/s41420-023-01772-9] [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: 05/24/2023] [Revised: 11/22/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024] Open
Abstract
The oncogenic MUC1-C transmembrane protein is a critical effector of the cancer stem cell (CSC) state. Addiction to MUC1-C for self-renewal in the progression of human cancers has emphasized the need for development of anti-MUC1-C agents. However, there are presently no approved small molecules for targeting MUC1-C-dependent CSCs. In screening for small molecules, we identified salinomycin (SAL), an inducer of ferroptosis, as a potent inhibitor of MUC1-C signaling. We demonstrate that SAL suppresses MUC1-C expression by disrupting a NF-κB/MUC1-C auto-inductive circuit that is necessary for ferroptosis resistance. Our results show that SAL-induced MUC1-C suppression downregulates a MUC1-C→MYC pathway that activates genes encoding (i) glutathione-disulfide reductase (GSR), and (ii) the LDL receptor related protein 8 (LRP8), which inhibit ferroptosis by generating GSH and regulating selenium levels, respectively. GSR and LRP8 contribute to the function of glutathione peroxidase 4 (GPX4), an essential negative regulator of ferroptotic cell death. We demonstrate that targeting MUC1-C genetically or with the GO-203 peptide inhibitor suppresses GPX4 expression and GPX activity in association with the induction of ferroptosis. Studies of CSCs enriched by serial passage as tumorspheres further demonstrate that the effects of SAL are mediated by downregulation of MUC1-C and thereby overcoming resistance to ferroptosis. As confirmation of these results, rescue of MUC1-C downregulation with the MUC1-C cytoplasmic domain (i) reversed the suppression of GSR, LRP8 and GPX4 expression, and (ii) attenuated the induction of ferroptosis. These findings identify SAL as a unique small molecule inhibitor of MUC1-C signaling and demonstrate that MUC1-C is an important effector of resistance to ferroptosis.
Collapse
Affiliation(s)
- Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Keyi Wang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Ayako Nakashoji
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hiroki Ozawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yoshihiro Morimoto
- Department of Gastroenterological Surgery, Kinan Hospital, Wakayama, Japan
| | - Nami Yamashita
- Breast Surgical Oncology, Breast Oncology Center, The Cancer Institute Hospital of the JFCR, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Shokrollahi Barough M, Jalili N, Shafiee S, Salehi M, Naseri N, Javidi MA, Hosseinzadeh A, Farahmand L. Anti-MUC1 nanobody can synergize the Tamoxifen and Herceptin effects on breast cancer cells by inducing ER, PR and HER2 overexpression. Int Immunopharmacol 2023; 124:110792. [PMID: 37633239 DOI: 10.1016/j.intimp.2023.110792] [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: 06/18/2023] [Accepted: 08/10/2023] [Indexed: 08/28/2023]
Abstract
INTRODUCTION One of the most pressing concerns associated with breast cancer-targeted therapies is resistance to Tamoxifen and Herceptin. Such drug resistance is usually characterized by reduced expression of certain cell surface receptors. Some biological regimens can induce perceptible overexpression of these receptors in favor of drug responsiveness. MATERIAL AND METHODS In this research, drug-responsive MCF-7 and SKBR-3, along with drug-resistant MCF-7R (Tamoxifen resistant) and JIMT-1 (Herceptin resistant) breast cancer cell lines in 2D and 3D cultures were exposed to anti-MUC1 nanobody and then assessed for their ER, PR, and HER2 gene and protein expression using qRT-PCR and immunofluorescent staining analyses. Cell viability and the synergistic relationships of combination treatments were determined with MTT assay followed by CompuSyn software. Apoptotic cells were evaluated with Annexin V/propidium Iodide (PI) and acridine orange/ethidium bromide (AO/EB) staining. RESULTS Anti-MUC1 exposure elevated the expression levels of ER (42 folds), PR (18.5 folds), and HER2 (4.7 folds). As a result of co-treatment, the IC50 levels for Tamoxifen and Herceptin were reduced by up to 10 and 3 folds, respectively. MCF-7R cells responded positively to Tamoxifen, as evidenced by a 5-fold reduction in the IC50 and enhanced apoptosis. CONCLUSION The ER, PR, and HER2 overexpression after MUC1 blocking could signal drug hypersensitization and facilitate drug resistance management.
Collapse
Affiliation(s)
- Mahdieh Shokrollahi Barough
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran; Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran; Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Neda Jalili
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Soodabe Shafiee
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Malihe Salehi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Nazanin Naseri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Aysooda Hosseinzadeh
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
3
|
Jin W, Zhang M, Dong C, Huang L, Luo Q. The multifaceted role of MUC1 in tumor therapy resistance. Clin Exp Med 2023; 23:1441-1474. [PMID: 36564679 DOI: 10.1007/s10238-022-00978-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
Tumor therapeutic resistances are frequently linked to the recurrence and poor prognosis of cancers and have been a key bottleneck in clinical tumor treatment. Mucin1 (MUC1), a heterodimeric transmembrane glycoprotein, exhibits abnormally overexpression in a variety of human tumors and has been confirmed to be related to the formation of therapeutic resistance. In this review, the multifaceted roles of MUC1 in tumor therapy resistance are summarized from aspects of pan-cancer principles shared among therapies and individual mechanisms dependent on different therapies. Concretely, the common mechanisms of therapy resistance across cancers include interfering with gene expression, promoting genome instability, modifying tumor microenvironment, enhancing cancer heterogeneity and stemness, and activating evasion and metastasis. Moreover, the individual mechanisms of therapy resistance in chemotherapy, radiotherapy, and biotherapy are introduced. Last but not least, MUC1-involved therapy resistance in different types of cancers and MUC1-related clinical trials are summarized.
Collapse
Affiliation(s)
- Weiqiu Jin
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Mengwei Zhang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Changzi Dong
- Department of Bioengineering, School of Engineering and Science, University of Pennsylvania, Philadelphia, 19104, USA
| | - Lei Huang
- Department of Histoembryology, Genetics and Developmental Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
4
|
Morimoto Y, Yamashita N, Hirose H, Fushimi A, Haratake N, Daimon T, Bhattacharya A, Ahmad R, Suzuki Y, Takahashi H, Kufe DW. MUC1-C is necessary for SHP2 activation and BRAF inhibitor resistance in BRAF(V600E) mutant colorectal cancer. Cancer Lett 2023; 559:216116. [PMID: 36878307 PMCID: PMC10408991 DOI: 10.1016/j.canlet.2023.216116] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/19/2023] [Accepted: 03/02/2023] [Indexed: 03/07/2023]
Abstract
Colorectal cancers (CRCs) harboring the BRAF(V600E) mutation are associated with aggressive disease and resistance to BRAF inhibitors by feedback activation of the receptor tyrosine kinase (RTK)→RAS→MAPK pathway. The oncogenic MUC1-C protein promotes progression of colitis to CRC; whereas there is no known involvement of MUC1-C in BRAF(V600E) CRCs. The present work demonstrates that MUC1 expression is significantly upregulated in BRAF(V600E) vs wild-type CRCs. We show that BRAF(V600E) CRC cells are dependent on MUC1-C for proliferation and BRAF inhibitor (BRAFi) resistance. Mechanistically, MUC1-C integrates induction of MYC in driving cell cycle progression with activation of the SHP2 phosphotyrosine phosphatase, which enhances RTK-mediated RAS→ERK signaling. We demonstrate that targeting MUC1-C genetically and pharmacologically suppresses (i) activation of MYC, (ii) induction of the NOTCH1 stemness factor, and (iii) the capacity for self-renewal. We also show that MUC1-C associates with SHP2 and is required for SHP2 activation in driving BRAFi-induced feedback of ERK signaling. In this way, targeting MUC1-C in BRAFi-resistant BRAF(V600E) CRC tumors inhibits growth and sensitizes to BRAF inhibition. These findings demonstrate that MUC1-C is a target for the treatment of BRAF(V600E) CRCs and for reversing their resistance to BRAF inhibitors by suppressing the feedback MAPK pathway.
Collapse
Affiliation(s)
| | - Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Haruka Hirose
- Division of Systems Biology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - Atsushi Fushimi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Naoki Haratake
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tatsuaki Daimon
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yozo Suzuki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Hidekazu Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Semba R, Horimoto Y, Sakata-Matsuzawa M, Ishizuka Y, Denda-Nagai K, Fujihira H, Noji M, Onagi H, Ichida M, Miura H, Watanabe J, Saito M, Saito T, Arakawa A, Irimura T. Possible correlation of apical localization of MUC1 glycoprotein with luminal A-like status of breast cancer. Sci Rep 2023; 13:5281. [PMID: 37002293 PMCID: PMC10066179 DOI: 10.1038/s41598-023-32579-4] [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: 09/21/2022] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Adjuvant chemotherapy has played a major role in the treatment of hormone receptor-positive breast cancer for many years. To better determine which patient subsets need adjuvant chemotherapy, various gene expression analyses have been developed, but cost-effective tools to identify such patients remain elusive. In the present report, we retrospectively investigated immunohistochemical expression and subcellular localization of MUC1 in primary tumors and examined their relationship to tumor malignancy, chemotherapy effect and patient outcomes. We retrospectively examined three patient cohorts with hormone receptor-positive/human epidermal growth factor receptor 2-negative invasive breast cancer: 51 patients who underwent 21-gene expression analysis (multi-gene assay-cohort), 96 patients who received neoadjuvant chemotherapy (neoadjuvant chemotherapy-cohort), and 609 patients whose tumor tissue was used in tissue-microarrays (tissue-microarray-cohort). The immunohistochemical staining pattern of the anti-MUC1 monoclonal antibody, Ma695, was examined in cancer tissues, and subcellular localization was determined as apical, cytoplasmic or negative. In the multi-gene assay-cohort, tumors with apical patterns had the lowest recurrence scores, reflecting lower tumor malignancy, and were significantly lower than MUC1-negative tumors (P = 0.038). In the neoadjuvant chemotherapy-cohort, there was no correlation between MUC1 staining patterns and effects of chemotherapy. Finally, in the tissue-microarray-cohort, we found that patients with apical MUC1 staining patterns had significantly longer disease-free-survival and overall survival than other patterns (P = 0.020 and 0.039, respectively). Our data suggest that an apical MUC1 staining pattern indicates luminal A-likeness. Assessment of the subcellular localization of MUC1 glycoprotein may be useful for identifying patients who can avoid adjuvant chemotherapy.
Collapse
Affiliation(s)
- Ryoko Semba
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Yoshiya Horimoto
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan.
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan.
| | - Madoka Sakata-Matsuzawa
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Yumiko Ishizuka
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Kaori Denda-Nagai
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Haruhiko Fujihira
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, RIKEN, Wako, Japan
| | - Miki Noji
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hiroko Onagi
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Miyu Ichida
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Hiroyoshi Miura
- Department of Surgery, Koshigaya Municipal Hospital, Saitama, Japan
| | - Junichiro Watanabe
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Mitsue Saito
- Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, 113-0033, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsushi Arakawa
- Department of Human Pathology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Tatsuro Irimura
- Division of Glycobiologics, Department of Breast Oncology, Juntendo University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Chen K, Xu J, Tong YL, Yan JF, Pan Y, Wang WJ, Zheng L, Zheng XX, Hu C, Hu X, Shen X, Chen W. Rab31 promotes metastasis and cisplatin resistance in stomach adenocarcinoma through Twist1-mediated EMT. Cell Death Dis 2023; 14:115. [PMID: 36781842 PMCID: PMC9925739 DOI: 10.1038/s41419-023-05596-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 02/15/2023]
Abstract
Stomach adenocarcinoma (STAD) is one of the leading causes of cancer-related death globally. Metastasis and drug resistance are two major causes of failures in current chemotherapy. Here, we found that the expression of Ras-related protein 31 (Rab31) is upregulated in human STAD tissues and high expression of Rab31 is closely associated with poor survival time. Furthermore, we revealed that Rab31 promotes cisplatin resistance and metastasis in human STAD cells. Reduced Rab31 expression induces tumor cell apoptosis and increases cisplatin sensitivity in STAD cells; Rab31 overexpression yielded the opposite result. Rab31 silencing prevented STAD cell migration, whereas the overexpression of Rab31 increased the metastatic potential. Further work showed that Rab31 mediates cisplatin resistance and metastasis via epithelial-mesenchymal transition (EMT) pathway. In addition, we found that both Rab31 overexpression and cisplatin treatment results in increased Twist1 expression. Depletion of Twist1 enhances sensitivity to cisplatin in STAD cells, which cannot be fully reversed by Rab31 overexpression. Rab31 could activate Twist1 by activating Stat3 and inhibiting Mucin 1 (MUC-1). The present study also demonstrates that Rab31 knockdown inhibited tumor growth in mice STAD models. These findings indicate that Rab31 is a novel and promising biomarker and potential therapeutic target for diagnosis, treatment and prognosis prediction in STAD patients. Our data not only identifies a novel Rab31/Stat3/MUC-1/Twist1/EMT pathway in STAD metastasis and drug resistance, but it also provides direction for the exploration of novel strategies to predict and treat STAD in the future.
Collapse
Affiliation(s)
- Ke Chen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Ji Xu
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Yu-Ling Tong
- Department of General Practice, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Jia-Fei Yan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Yu Pan
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang Province, China
| | - Wei-Jia Wang
- Department of Pharmacy, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang Province, China
| | - Li Zheng
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang Province, China
| | - Xiao-Xiao Zheng
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang Province, China
| | - Can Hu
- Department of Gastric Surgery, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang Province, China
| | - Xiu Hu
- Department of Pharmacy, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang Province, China.
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China.
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang Province, China.
- Institute of Clinical Medicine Research, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China.
| |
Collapse
|
7
|
Xu X, Li Y, Liu G, Li K, Chen P, Gao Y, Liang W, Xi H, Wang X, Wei B, Li H, Chen L. MiR-378a-3p acts as a tumor suppressor in gastric cancer via directly targeting RAB31 and inhibiting the Hedgehog pathway proteins GLI1/2. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0337. [PMID: 36245214 PMCID: PMC9755959 DOI: 10.20892/j.issn.2095-3941.2022.0337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE To improve the prognosis of patients with gastric cancer (GC), more effective therapeutic targets are urgently needed. Increasing evidence indicates that miRNAs are involved in the progression of various tumors, and RAS-associated protein in the brain 31 (RAB31) is upregulated and promotes the progression of multiple malignant tumors. Here, we focused on identifying RAB31-targeted miRNAs and elucidating their potential mechanism in the progression of GC. METHODS RAB31 and miR-378a-3p expression levels were detected in paired fresh GC tissues and GC cell lines. Bioinformatics analysis was used to predict the miRNAs targeting RAB31 and the relationships between RAB31 and other genes. Dual-luciferase reporter assays were applied to verify the targeted interaction relationship. CCK-8, colony formation, flow cytometry, wound healing, and Transwell assays were performed to assess the proliferation, apoptosis, migration, and invasion of GC cells. Tumorsphere formation assays were performed to assess the stemness of gastric cancer stem cells. Related proteins were detected by Western blot. Xenograft assays in nude mice were performed to explore the effect of miR-378a-3p in vivo. RESULTS We report the first evidence that miR-378a-3p is downregulated in GC, whereas its overexpression inhibits proliferation, invasion, and migration as well as promotes apoptosis in GC cells. Mechanistically, miR-378a-3p inhibits the progression of GC by directly targeting RAB31. Restoring RAB31 expression partially offsets the inhibitory effect of miR-378a-3p. Further research revealed that miR-378a-3p inhibits GLI1/2 in the Hedgehog signaling pathway and attenuates the stemness of gastric cancer stem cells. Finally, xenograft assays showed that miR-378a-3p inhibits GC tumorigenesis in vivo. CONCLUSIONS MiR-378a-3p inhibits GC progression by directly targeting RAB31 and inhibiting the Hedgehog signaling pathway proteins GLI1/2.
Collapse
Affiliation(s)
- Xinxin Xu
- Medical School of Chinese PLA, Beijing 100853, China,Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yang Li
- Medical School of Chinese PLA, Beijing 100853, China,Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Guoxiao Liu
- Medical School of Chinese PLA, Beijing 100853, China,Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Kai Li
- Medical School of Chinese PLA, Beijing 100853, China,Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Peng Chen
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of People’s Liberation Army, Lanzhou 730050, China
| | - Yunhe Gao
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Wenquan Liang
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongqing Xi
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xinxin Wang
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Bo Wei
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongtao Li
- Department of General Surgery, The 940th Hospital of Joint Logistics Support Force of People’s Liberation Army, Lanzhou 730050, China,Correspondence to: Hongtao Li and Lin Chen, E-mail: and
| | - Lin Chen
- Senior Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China,Correspondence to: Hongtao Li and Lin Chen, E-mail: and
| |
Collapse
|
8
|
Yamashita N, Kufe D. Addiction of Cancer Stem Cells to MUC1-C in Triple-Negative Breast Cancer Progression. Int J Mol Sci 2022; 23:8219. [PMID: 35897789 PMCID: PMC9331006 DOI: 10.3390/ijms23158219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 02/01/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy with limited treatment options. TNBC progression is associated with expansion of cancer stem cells (CSCs). Few insights are available regarding druggable targets that drive the TNBC CSC state. This review summarizes the literature on TNBC CSCs and the compelling evidence that they are addicted to the MUC1-C transmembrane protein. In normal epithelia, MUC1-C is activated by loss of homeostasis and induces reversible wound-healing responses of inflammation and repair. However, in settings of chronic inflammation, MUC1-C promotes carcinogenesis. MUC1-C induces EMT, epigenetic reprogramming and chromatin remodeling in TNBC CSCs, which are dependent on MUC1-C for self-renewal and tumorigenicity. MUC1-C-induced lineage plasticity in TNBC CSCs confers DNA damage resistance and immune evasion by chronic activation of inflammatory pathways and global changes in chromatin architecture. Of therapeutic significance, an antibody generated against the MUC1-C extracellular domain has been advanced in a clinical trial of anti-MUC1-C CAR T cells and in IND-enabling studies for development as an antibody-drug conjugate (ADC). Agents targeting the MUC1-C cytoplasmic domain have also entered the clinic and are undergoing further development as candidates for advancing TNBC treatment. Eliminating TNBC CSCs will be necessary for curing this recalcitrant cancer and MUC1-C represents a promising druggable target for achieving that goal.
Collapse
Affiliation(s)
- Nami Yamashita
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
9
|
Hosseinzadeh A, Merikhian P, Naseri N, Eisavand MR, Farahmand L. MUC1 is a potential target to overcome trastuzumab resistance in breast cancer therapy. Cancer Cell Int 2022; 22:110. [PMID: 35248049 PMCID: PMC8897942 DOI: 10.1186/s12935-022-02523-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/12/2022] [Indexed: 02/07/2023] Open
Abstract
Although resistance is its major obstacle in cancer therapy, trastuzumab is the most successful agent in treating epidermal growth factor receptor 2 positive (HER2 +) breast cancer (BC). Some patients show resistance to trastuzumab, and scientists want to circumvent this problem. This review elaborately discusses possible resistance mechanisms to trastuzumab and introduces mucin 1 (MUC1) as a potential target efficient for overcoming such resistance. MUC1 belongs to the mucin family, playing the oncogenic/mitogenic roles in cancer cells and interacting with several other oncogenic receptors and pathways, such as HER2, β-catenin, NF-κB, and estrogen receptor (ERα). Besides, it has been established that MUC1- Cytoplasmic Domain (MUC1-CD) accelerates the development of resistance to trastuzumab and that silencing MUC1-C proto-oncogene is associated with increased sensitivity of HER2+ cells to trastuzumab-induced growth inhibitors. We mention why targeting MUC1 can be useful in overcoming trastuzumab resistance in cancer therapy.
Collapse
|
10
|
Kufe DW. MUC1-C in chronic inflammation and carcinogenesis; emergence as a target for cancer treatment. Carcinogenesis 2021; 41:1173-1183. [PMID: 32710608 DOI: 10.1093/carcin/bgaa082] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/14/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic inflammation is a highly prevalent consequence of changes in environmental and lifestyle factors that contribute to the development of cancer. The basis for this critical association has largely remained unclear. The MUC1 gene evolved in mammals to protect epithelia from the external environment. The MUC1-C subunit promotes responses found in wound healing and cancer. MUC1-C induces EMT, epigenetic reprogramming, dedifferentiation and pluripotency factor expression, which when prolonged in chronic inflammation promote cancer progression. As discussed in this review, MUC1-C also drives drug resistance and immune evasion, and is an important target for cancer therapeutics now under development.
Collapse
Affiliation(s)
- Donald W Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Jahangiri R, Mosaffa F, Emami Razavi A, Teimoori-Toolabi L, Jamialahmadi K. PAX2 promoter methylation and AIB1 overexpression promote tamoxifen resistance in breast carcinoma patients. J Oncol Pharm Pract 2021; 28:310-325. [PMID: 33509057 DOI: 10.1177/1078155221989404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Disease recurrence is an important obstacle in estrogen receptor positive (ER+) tamoxifen treated breast carcinoma patients. Tamoxifen resistance-related molecular mechanisms are not fully understood. Alteration in DNA methylation which contributes to transcriptional regulation of cancer-related genes plays a crucial role in tamoxifen response. In the present study, the contribution of promoter methylation and mRNA expression of PAX2 and AIB1 in the development of breast carcinoma and tamoxifen refractory was assessed. METHODS Methylation specific-high resolution melting (MS-HRM) analysis and Real-time quantitative PCR (RT-qPCR) experiment were performed to analyze the promoter methylation and mRNA expression levels of PAX2 and AIB1 genes in 102 breast tumors and adjacent normal breast specimens. RESULTS We indicated that PAX2 expression is decreased in breast tissues due to hypermethylation in its promoter region. Compared to the adjacent normal tissues, the tumors exhibited significantly lower relative mRNA levels of PAX2 and increased expression of AIB1. Aberrant promoter methylation of PAX2 and overexpression of AIB1 was observed in tamoxifen resistance patients compared to the sensitive ones. Cox regression analysis exhibited that the increased promoter methylation status of PAX2 and overexpression of AIB1 remained as unfavorable identifiers which influence patients' survival independently. CONCLUSIONS Our results revealed that the aberration in PAX2 promoter methylation and AIB1 overexpression are associated with the tamoxifen response in breast carcinoma patients. Further research is needed to demonstrate the potential of using PAX2 and AIB1 expression and their methylation-mediated regulation as predictive or prognostic biomarkers or as a new target therapy for better disease management.
Collapse
Affiliation(s)
- Rosa Jahangiri
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mosaffa
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | | | - Khadijeh Jamialahmadi
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Abdoli Shadbad M, Hajiasgharzadeh K, Baradaran B. Cross-talk between myeloid-derived suppressor cells and Mucin1 in breast cancer vaccination: On the verge of a breakthrough. Life Sci 2020; 258:118128. [PMID: 32710947 DOI: 10.1016/j.lfs.2020.118128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/16/2020] [Accepted: 07/17/2020] [Indexed: 01/22/2023]
Abstract
Although breast cancer is one of the leading troublesome cancers, the available therapeutic options have not fulfilled the desired outcomes. Immune-based therapy has gained special attention for breast cancer treatment. Although this approach is highly tolerable, its low response rate has rendered it as an undesirable approach. This review aims to describe the essential oncogenic pathways involved in breast cancer, elucidate the immunosuppression and oncogenic effect of Mucin1, and introduce myeloid-derived suppressor cells, which are the main culprits of anti-tumoral immune response attenuation. The various auto-inductive loops between Mucin1 and myeloid-derived suppressor cells are focal in the suppression of anti-tumoral immune responses in patients with breast cancer. These cross-talks between the Mucin1 and myeloid-derived suppressor cells can be the underlying causes of immunotherapy's impotence for patients with breast cancer. This approach can pave the road for the development of a potent vaccine for patients with breast cancer and is translated into clinical settings.
Collapse
Affiliation(s)
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
13
|
Simões BM, Santiago-Gómez A, Chiodo C, Moreira T, Conole D, Lovell S, Alferez D, Eyre R, Spence K, Sarmiento-Castro A, Kohler B, Morisset L, Lanzino M, Andò S, Marangoni E, Sims AH, Tate EW, Howell SJ, Clarke RB. Targeting STAT3 signaling using stabilised sulforaphane (SFX-01) inhibits endocrine resistant stem-like cells in ER-positive breast cancer. Oncogene 2020; 39:4896-4908. [PMID: 32472077 PMCID: PMC7299846 DOI: 10.1038/s41388-020-1335-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 01/05/2023]
Abstract
Estrogen receptor (ER) positive breast cancer is frequently sensitive to endocrine therapy. Multiple mechanisms of endocrine therapy resistance have been identified, including cancer stem-like cell (CSC) activity. Here we investigate SFX-01, a stabilised formulation of sulforaphane (SFN), for its effects on breast CSC activity in ER+ preclinical models. SFX-01 reduced mammosphere formation efficiency (MFE) of ER+ primary and metastatic patient samples. Both tamoxifen and fulvestrant increased MFE and aldehyde dehydrogenase (ALDH) activity of patient-derived xenograft (PDX) tumors, which was reversed by combination with SFX-01. SFX-01 significantly reduced tumor-initiating cell frequency in secondary transplants and reduced the formation of spontaneous lung micrometastases by PDX tumors in mice. Mechanistically, we establish that both tamoxifen and fulvestrant induce STAT3 phosphorylation. SFX-01 suppressed phospho-STAT3 and SFN directly bound STAT3 in patient and PDX samples. Analysis of ALDH+ cells from endocrine-resistant patient samples revealed activation of STAT3 target genes MUC1 and OSMR, which were inhibited by SFX-01 in patient samples. Increased expression of these genes after 3 months' endocrine treatment of ER+ patients (n = 68) predicted poor prognosis. Our data establish the importance of STAT3 signaling in CSC-mediated resistance to endocrine therapy and the potential of SFX-01 for improving clinical outcomes in ER+ breast cancer.
Collapse
Affiliation(s)
- Bruno M Simões
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Angélica Santiago-Gómez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Chiara Chiodo
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Pharmacy, University of Calabria, Arcavacata di Rende, Italy
| | - Tiago Moreira
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Daniel Conole
- Molecular Sciences Research Hub, Imperial College, London, UK
| | - Scott Lovell
- Molecular Sciences Research Hub, Imperial College, London, UK
| | - Denis Alferez
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rachel Eyre
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Katherine Spence
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Aida Sarmiento-Castro
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Bertram Kohler
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ludivine Morisset
- Institut Curie, PSL Research University, Translational Research Department, Paris, France
| | - Marilena Lanzino
- Department of Pharmacy, University of Calabria, Arcavacata di Rende, Italy
| | - Sebastiano Andò
- Department of Pharmacy, University of Calabria, Arcavacata di Rende, Italy
| | - Elisabetta Marangoni
- Institut Curie, PSL Research University, Translational Research Department, Paris, France
| | - Andrew H Sims
- Applied Bioinformatics of Cancer Group, University of Edinburgh Cancer Research UK Centre, Edinburgh, UK
| | - Edward W Tate
- Molecular Sciences Research Hub, Imperial College, London, UK
| | - Sacha J Howell
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.
| | - Robert B Clarke
- Breast Biology Group, Manchester Breast Centre, Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
14
|
Reynolds IS, Fichtner M, McNamara DA, Kay EW, Prehn JHM, Burke JP. Mucin glycoproteins block apoptosis; promote invasion, proliferation, and migration; and cause chemoresistance through diverse pathways in epithelial cancers. Cancer Metastasis Rev 2020; 38:237-257. [PMID: 30680581 DOI: 10.1007/s10555-019-09781-w] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Overexpression of mucin glycoproteins has been demonstrated in many epithelial-derived cancers. The significance of this overexpression remains uncertain. The aim of this paper was to define the association of mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers by performing a systematic review of all published data. A systematic review of PubMed, Embase, and the Cochrane Central Register of Controlled Trials was performed to identify all papers that evaluated the association between mucin glycoproteins with apoptosis, cell growth, invasion, migration, adhesion, and clonogenicity in vitro as well as tumor growth, tumorigenicity, and metastasis in vivo in epithelial-derived cancers. PRISMA guidelines were adhered to. Results of individual studies were extracted and pooled together based on the organ in which the cancer was derived from. The initial search revealed 2031 papers, of which 90 were deemed eligible for inclusion in the study. The studies included details on MUC1, MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16. The majority of studies evaluated MUC1. MUC1 overexpression was consistently associated with resistance to apoptosis and resistance to chemotherapy. There was also evidence that overexpression of MUC2, MUC4, MUC5AC, MUC5B, MUC13, and MUC16 conferred resistance to apoptosis in epithelial-derived cancers. The overexpression of mucin glycoproteins is associated with resistance to apoptosis in numerous epithelial cancers. They cause resistance through diverse signaling pathways. Targeting the expression of mucin glycoproteins represents a potential therapeutic target in the treatment of epithelial-derived cancers.
Collapse
Affiliation(s)
- Ian S Reynolds
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Michael Fichtner
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Deborah A McNamara
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland
- Department of Surgery, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Elaine W Kay
- Department of Pathology, Beaumont Hospital, Dublin 9, Ireland
- Department of Pathology, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephens Green, Dublin 2, Ireland
| | - John P Burke
- Department of Colorectal Surgery, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
15
|
Gatti‐Mays ME, Redman JM, Donahue RN, Palena C, Madan RA, Karzai F, Bilusic M, Sater HA, Marté JL, Cordes LM, McMahon S, Steinberg SM, Orpia A, Burmeister A, Schlom J, Gulley JL, Strauss J. A Phase I Trial Using a Multitargeted Recombinant Adenovirus 5 (CEA/MUC1/Brachyury)-Based Immunotherapy Vaccine Regimen in Patients with Advanced Cancer. Oncologist 2019; 25:479-e899. [PMID: 31594913 PMCID: PMC7288633 DOI: 10.1634/theoncologist.2019-0608] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/05/2019] [Indexed: 12/31/2022] Open
Abstract
LESSONS LEARNED Concurrent ETBX-011, ETBX-051, and ETBX-061 can be safely administered to patients with advanced cancer. All patients developed CD4+ and/or CD8+ T-cell responses after vaccination to at least one tumor-associated antigen (TAA) encoded by the vaccine; 5/6 patients (83%) developed MUC1-specific T cells, 4/6 (67%) developed CEA-specific T cells, and 3/6 (50%) developed brachyury-specific T cells. The presence of adenovirus 5-neutralizing antibodies did not prevent the generation of TAA-specific T cells. BACKGROUND A novel adenovirus-based vaccine targeting three human tumor-associated antigens-CEA, MUC1, and brachyury-has demonstrated antitumor cytolytic T-cell responses in preclinical animal models of cancer. METHODS This open-label, phase I trial evaluated concurrent administration of three therapeutic vaccines (ETBX-011 = CEA, ETBX-061 = MUC1 and ETBX-051 = brachyury). All three vaccines used the same modified adenovirus 5 (Ad5) vector backbone and were administered at a single dose level (DL) of 5 × 1011 viral particles (VP) per vector. The vaccine regimen consisting of all three vaccines was given every 3 weeks for three doses then every 8 weeks for up to 1 year. Clinical and immune responses were evaluated. RESULTS Ten patients enrolled on trial (DL1 = 6 with 4 in the DL1 expansion cohort). All treatment-related adverse events were temporary, self-limiting, grade 1/2 and included injection site reactions and flu-like symptoms. Antigen-specific T cells to MUC1, CEA, and/or brachyury were generated in all patients. There was no evidence of antigenic competition. The administration of the vaccine regimen produced stable disease as the best clinical response. CONCLUSION Concurrent ETBX-011, ETBX-051, and ETBX-061 can be safely administered to patients with advanced cancer. Further studies of the vaccine regimen in combination with other agents, including immune checkpoint blockade, are planned.
Collapse
Affiliation(s)
- Margaret E. Gatti‐Mays
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jason M. Redman
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Renee N. Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Ravi A. Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Fatima Karzai
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Marijo Bilusic
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Houssein Abdul Sater
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Jennifer L. Marté
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Lisa M. Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Sheri McMahon
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Alanvin Orpia
- Leidos Biomedical Research, Inc.FrederickMarylandUSA
| | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - James L. Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Julius Strauss
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
16
|
Hata T, Rajabi H, Yamamoto M, Jin C, Ahmad R, Zhang Y, Kui L, Li W, Yasumizu Y, Hong D, Miyo M, Hiraki M, Maeda T, Suzuki Y, Takahashi H, Samur M, Kufe D. Targeting MUC1-C Inhibits TWIST1 Signaling in Triple-Negative Breast Cancer. Mol Cancer Ther 2019; 18:1744-1754. [PMID: 31308076 DOI: 10.1158/1535-7163.mct-19-0156] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/21/2019] [Accepted: 07/08/2019] [Indexed: 01/06/2023]
Abstract
The oncogenic MUC1-C protein and the TWIST1 epithelial-mesenchymal transition transcription factor (EMT-TF) are aberrantly expressed in triple-negative breast cancer (TNBC) cells. However, there is no known association between MUC1-C and TWIST1 in TNBC or other cancer cells. Here, we show that MUC1-C activates STAT3, and that MUC1-C and pSTAT3 drive induction of the TWIST1 gene. In turn, MUC1-C binds directly to TWIST1, and MUC1-C/TWIST1 complexes activate MUC1-C expression in an autoinductive circuit. The functional significance of the MUC1-C/TWIST1 circuit is supported by the demonstration that this pathway is sufficient for driving (i) the EMT-TFs, ZEB1 and SNAIL, (ii) multiple genes in the EMT program as determined by RNA-seq, and (iii) the capacity for cell invasion. We also demonstrate that the MUC1-C/TWIST1 circuit drives (i) expression of the stem cell markers SOX2, BMI1, ALDH1, and CD44, (ii) self-renewal capacity, and (iii) tumorigenicity. In concert with these results, we show that MUC1-C and TWIST1 also drive EMT and stemness in association with acquired paclitaxel (PTX) resistance. Of potential therapeutic importance, targeting MUC1-C and thereby TWIST1 reverses the PTX refractory phenotype as evidenced by synergistic activity with PTX against drug-resistant cells. These findings uncover a master role for MUC1-C in driving the induction of TWIST1, EMT, stemness, and drug resistance, and support MUC1-C as a highly attractive target for inhibiting TNBC plasticity and progression.
Collapse
Affiliation(s)
- Tsuyoshi Hata
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Yamamoto
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Caining Jin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yan Zhang
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ling Kui
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Wei Li
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yota Yasumizu
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Deli Hong
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masaaki Miyo
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Masayuki Hiraki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Takahiro Maeda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yozo Suzuki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hidekazu Takahashi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mehmet Samur
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
17
|
Linares A, Assou S, Lapierre M, Thouennon E, Duraffourd C, Fromaget C, Boulahtouf A, Tian G, Ji J, Sahin O, Badia E, Boulle N, Cavaillès V. Increased expression of the HDAC9 gene is associated with antiestrogen resistance of breast cancers. Mol Oncol 2019; 13:1534-1547. [PMID: 31099456 PMCID: PMC6599838 DOI: 10.1002/1878-0261.12505] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 04/26/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
Estrogens play a pivotal role in breast cancer etiology, and endocrine therapy remains the main first line treatment for estrogen receptor‐alpha (ERα)‐positive breast cancer. ER are transcription factors whose activity is finely regulated by various regulatory complexes, including histone deacetylases (HDACs). Here, we investigated the role of HDAC9 in ERα signaling and response to antiestrogens in breast cancer cells. Various Michigan Cancer Foundation‐7 (MCF7) breast cancer cell lines that overexpress class IIa HDAC9 or that are resistant to the partial antiestrogen 4‐hydroxy‐tamoxifen (OHTam) were used to study phenotypic changes in response to ER ligands by using transcriptomic and gene set enrichment analyses. Kaplan–Meier survival analyses were performed using public transcriptomic datasets from human breast cancer biopsies. In MCF7 breast cancer cells, HDAC9 decreased ERα mRNA and protein expression and inhibited its transcriptional activity. Conversely, HDAC9 mRNA was strongly overexpressed in OHTam‐resistant MCF7 cells and in ERα‐negative breast tumor cell lines. Moreover, HDAC9‐overexpressing cells were less sensitive to OHTam antiproliferative effects compared with parental MCF7 cells. Several genes (including MUC1, SMC3 and S100P) were similarly deregulated in OHTam‐resistant and in HDAC9‐overexpressing MCF7 cells. Finally, HDAC9 expression was positively associated with genes upregulated in endocrine therapy‐resistant breast cancers and high HDAC9 levels were associated with worse prognosis in patients treated with OHTam. These results demonstrate the complex interactions of class IIa HDAC9 with ERα signaling in breast cancer cells and its effect on the response to hormone therapy.
Collapse
Affiliation(s)
- Aurélien Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Said Assou
- Université Montpellier, France.,IRMB, Institute for Regenerative Medicine & Biotherapy, Montpellier, France.,INSERM, U1183, Montpellier, France
| | - Marion Lapierre
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Erwan Thouennon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Céline Duraffourd
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Carole Fromaget
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Abdelhay Boulahtouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Gao Tian
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, USA
| | - Eric Badia
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Nathalie Boulle
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France.,Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| |
Collapse
|
18
|
Gatti-Mays ME, Strauss J, Donahue RN, Palena C, Del Rivero J, Redman JM, Madan RA, Marté JL, Cordes LM, Lamping E, Orpia A, Burmeister A, Wagner E, Pico Navarro C, Heery CR, Schlom J, Gulley JL. A Phase I Dose-Escalation Trial of BN-CV301, a Recombinant Poxviral Vaccine Targeting MUC1 and CEA with Costimulatory Molecules. Clin Cancer Res 2019; 25:4933-4944. [PMID: 31110074 DOI: 10.1158/1078-0432.ccr-19-0183] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/26/2019] [Accepted: 05/16/2019] [Indexed: 01/28/2023]
Abstract
PURPOSE BN-CV301 is a poxviral-based vaccine comprised of recombinant (rec.) modified vaccinia Ankara (MVA-BN-CV301; prime) and rec. fowlpox (FPV-CV301; boost). Like its predecessor PANVAC, BN-CV301 contains transgenes encoding tumor-associated antigens MUC1 and CEA as well as costimulatory molecules (B7.1, ICAM-1, and LFA-3). PANVAC was reengineered to make it safer and more antigenic. PATIENTS AND METHODS This open-label, 3+3 design, dose-escalation trial evaluated three dose levels (DL) of MVA-BN-CV301: one, two, or four subcutaneous injections of 4 × 108 infectious units (Inf.U)/0.5 mL on weeks 0 and 4. All patients received FPV-CV301 subcutaneously at 1 × 109 Inf.U/0.5 mL every 2 weeks for 4 doses, then every 4 weeks. Clinical and immune responses were evaluated. RESULTS There were no dose-limiting toxicities. Twelve patients enrolled on trial [dose level (DL) 1 = 3, DL2 = 3, DL3 = 6). Most side effects were seen with the prime doses and lessened with subsequent boosters. All treatment-related adverse events were temporary, self-limiting, grade 1/2, and included injection-site reactions and flu-like symptoms. Antigen-specific T cells to MUC1 and CEA, as well as to a cascade antigen, brachyury, were generated in most patients. Single-agent BN-CV301 produced a confirmed partial response (PR) in 1 patient and prolonged stable disease (SD) in multiple patients, most notably in KRAS-mutant gastrointestinal tumors. Furthermore, 2 patients with KRAS-mutant colorectal cancer had prolonged SD when treated with an anti-PD-L1 antibody following BN-CV301. CONCLUSIONS The BN-CV301 vaccine can be safely administered to patients with advanced cancer. Further studies of the vaccine in combination with other agents are planned.See related commentary by Repáraz et al., p. 4871.
Collapse
Affiliation(s)
- Margaret E Gatti-Mays
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Julius Strauss
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Renee N Donahue
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jaydira Del Rivero
- Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason M Redman
- Medical Oncology Service, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ravi A Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L Marté
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa M Cordes
- Oncology Clinical Pharmacy, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Lamping
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Alanvin Orpia
- Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - Eva Wagner
- Bavarian Nordic GmbH, Martinsried, Germany
| | | | | | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
19
|
Heublein S, Page S, Mayr D, Schmoeckel E, Trillsch F, Marmé F, Mahner S, Jeschke U, Vattai A. Potential Interplay of the Gatipotuzumab Epitope TA-MUC1 and Estrogen Receptors in Ovarian Cancer. Int J Mol Sci 2019; 20:ijms20020295. [PMID: 30642093 PMCID: PMC6359481 DOI: 10.3390/ijms20020295] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/17/2018] [Accepted: 12/24/2018] [Indexed: 12/20/2022] Open
Abstract
Anti-tumor efficacy of Gatipotuzumab, a therapeutic antibody targeting Tumor-Associated Mucin-1 (TA-MUC1), in relapsed ovarian cancer (OC) appeared to be rather heterogeneous. Whether adding a second anti-neoplastic drug may augment response towards Gatipotuzumab, has not been elucidated so far. Since it is known that anti-MUC1 antibodies may alter estrogen receptor activity in breast cancer, this potential interplay was investigated in OC. The correlation between TA-MUC1, estrogen receptors (ERs) and another 12 protein markers as well as their correlation with clinico-pathological parameters in 138 ovarian cancer cases was studied. Finally, Gatipotuzumab and 4-Hydroxy-TTamoxifen (4-OHT) as well as the combination of both was tested for its impact on cell viability in COV318, OV-90, OVCAR-3, and SKOV-3 cells. A strong positive correlation between TA-MUC1 and ERs was detected in OC tissue. Those cases missing ERs but staining positive for TA-MUC1 had significantly reduced overall survival. The combination of 4-OHT and Gatipotuzumab significantly reduced cell viability and was more effective than treatment with Gatipotuzumab alone. Co-stimulation with Gatipotuzumab enhanced the efficacy of 4-OHT in OVCAR-3 and SKOV-3. The data suggest an interplay of TA-MUC1 and ERs in OC. Whether the combination of Gatipotuzumab and TTamoxifen may enhance efficacy of either of the two drugs in vivo, or may even translate into a clinically relevant benefit over the respective monotherapies, remains to be investigated.
Collapse
Affiliation(s)
- Sabine Heublein
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
- Department of Obstetrics and Gynecology, University of Heidelberg, 69117 Heidelberg, Germany.
| | - Sabina Page
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians University of Munich, 81377 Munich, Germany.
| | - Elisa Schmoeckel
- Department of Pathology, Ludwig-Maximilians University of Munich, 81377 Munich, Germany.
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University of Heidelberg, 69117 Heidelberg, Germany.
| | - Sven Mahner
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
| | - Aurelia Vattai
- Department of Obstetrics and Gynecology, Ludwig-Maximilians University of Munich, University Hospital, 81377 Munich, Germany.
| |
Collapse
|
20
|
Tang CT, Liang Q, Yang L, Lin XL, Wu S, Chen Y, Zhang XT, Gao YJ, Ge ZZ. RAB31 Targeted by MiR-30c-2-3p Regulates the GLI1 Signaling Pathway, Affecting Gastric Cancer Cell Proliferation and Apoptosis. Front Oncol 2018; 8:554. [PMID: 30534536 PMCID: PMC6275292 DOI: 10.3389/fonc.2018.00554] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 11/08/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Gastric cancer (GC), one of the most common cancers worldwide, is highly malignant and fatal. Ras-related protein in brain 31 (RAB31), a member of the RAB family of oncogenes, participates in the process of carcinogenesis and cancer development; however, its role in GC progression is unknown. Methods: In our study, 90 pairs of tissue microarrays were used to measure the levels of RAB31 protein by immunochemistry, and 22 pairs of fresh tissue were used to measure the levels of RAB31 mRNA by quantitative PCR. We also investigated the effects of RAB31 on tumor growth both in vitro and in vivo. Results: RAB31 was overexpressed in GC tissues, and its overexpression predicted poor survival in patients. In a nude mouse model, depletion of RAB31 inhibited tumor growth. In vitro, silencing of RAB31 suppressed cell viability, promoted cell cycle arrest, enhanced apoptosis, and affected the expression of cell cycle and apoptotic proteins; these effects were mediated by glioma-associated oncogene homolog 1 (GLI1). Co-immunoprecipitation and immunofluorescence assays confirmed that RAB31 interacted with GLI1. In addition, luciferase reporter assays and Western blotting showed that microRNA-30c-2-3p modulated the RAB31/GLI1 pathway by targeting the 3′-untranslated region of RAB31. Conclusions: Collectively, these data show that RAB31 is regulated by microRNA-30c-2-3p, and functions as an oncogene in GC tumorigenesis and development by interacting with GLI1. Therefore, targeting the miR-30c-2-3p/RAB31/GLI1 axis may be a therapeutic intervention for gastric cancer.
Collapse
Affiliation(s)
- Chao-Tao Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Liang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Yang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Lu Lin
- Department of Digestive Endoscopy, Provincial Clinic Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou, China
| | - Shan Wu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin-Tian Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yun-Jie Gao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Zheng Ge
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
21
|
Tecalco-Cruz AC, Ramírez-Jarquín JO. Polyubiquitination inhibition of estrogen receptor alpha and its implications in breast cancer. World J Clin Oncol 2018; 9:60-70. [PMID: 30148069 PMCID: PMC6107474 DOI: 10.5306/wjco.v9.i4.60] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/22/2018] [Accepted: 06/28/2018] [Indexed: 02/06/2023] Open
Abstract
Estrogen receptor alpha (ERα) is detected in more than 70% of the cases of breast cancer. Nuclear activity of ERα, a transcriptional regulator, is linked to the development of mammary tumors, whereas the extranuclear activity of ERα is related to endocrine therapy resistance. ERα polyubiquitination is induced by the estradiol hormone, and also by selective estrogen receptor degraders, resulting in ERα degradation via the ubiquitin proteasome system. Moreover, polyubiquitination is related to the ERα transcription cycle, and some E3-ubiquitin ligases also function as coactivators for ERα. Several studies have demonstrated that ERα polyubiquitination is inhibited by multiple mechanisms that include posttranslational modifications, interactions with coregulators, and formation of specific protein complexes with ERα. These events are responsible for an increase in ERα protein levels and deregulation of its signaling in breast cancers. Thus, ERα polyubiquitination inhibition may be a key factor in the progression of breast cancer and resistance to endocrine therapy.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Programa de Investigación de Cáncer de Mama (PICM), Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México 04510, México
| | - Josué O Ramírez-Jarquín
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México 04510, México
| |
Collapse
|
22
|
Jahangiri R, Mosaffa F, Gharib M, Emami Razavi AN, Abdirad A, Jamialahmadi K. PAX2 expression is correlated with better survival in tamoxifen-treated breast carcinoma patients. Tissue Cell 2018; 52:135-142. [DOI: 10.1016/j.tice.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/07/2018] [Indexed: 02/07/2023]
|
23
|
Rajabi H, Hiraki M, Kufe D. MUC1-C activates polycomb repressive complexes and downregulates tumor suppressor genes in human cancer cells. Oncogene 2018; 37:2079-2088. [PMID: 29379165 PMCID: PMC5908737 DOI: 10.1038/s41388-017-0096-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/19/2017] [Accepted: 09/29/2017] [Indexed: 12/11/2022]
Abstract
The PRC2 and PRC1 complexes are aberrantly expressed in human cancers and have been linked to decreases in patient survival. MUC1-C is an oncoprotein that is also overexpressed in diverse human cancers and is associated with a poor prognosis. Recent studies have supported a previously unreported function for MUC1-C in activating PRC2 and PRC1 in cancer cells. In the regulation of PRC2, MUC1-C (i) drives transcription of the EZH2 gene, (ii) binds directly to EZH2, and (iii) enhances occupancy of EZH2 on target gene promoters with an increase in H3K27 trimethylation. Regarding PRC1, which is recruited to PRC2 sites in the hierarchical model, MUC1-C induces BMI1 transcription, forms a complex with BMI1, and promotes H2A ubiquitylation. MUC1-C thereby contributes to the integration of PRC2 and PRC1-mediated repression of tumor suppressor genes, such as CDH1, CDKN2A, PTEN and BRCA1. Like PRC2 and PRC1, MUC1-C is associated with the epithelial-mesenchymal transition (EMT) program, cancer stem cell (CSC) state, and acquisition of anticancer drug resistance. In concert with these observations, targeting MUC1-C downregulates EZH2 and BMI1, inhibits EMT and the CSC state, and reverses drug resistance. These findings emphasize the significance of MUC1-C as a therapeutic target for inhibiting aberrant PRC function and reprogramming the epigenome in human cancers.
Collapse
Affiliation(s)
- Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Masayuki Hiraki
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
24
|
Yi FT, Lu QP. Mucin 1 promotes radioresistance in hepatocellular carcinoma cells through activation of JAK2/STAT3 signaling. Oncol Lett 2017; 14:7571-7576. [PMID: 29344203 DOI: 10.3892/ol.2017.7119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/14/2017] [Indexed: 12/15/2022] Open
Abstract
Mucin 1 (MUC1) is aberrantly overexpressed in numerous human cancer types, including hepatocellular carcinoma (HCC) and contributes to chemoresistance of tumor cells. The aim of the present study was to evaluate the possible implication of MUC1 in radioresistance of HCC cells and the underlying mechanisms. It was demonstrated that MUC1 was significantly upregulated in HCC cells following irradiation exposure, which was coupled with increased phosphorylation of signal transducer and activator of transcription 3 (STAT3). Enforced expression of MUC1 significantly (P<0.05) promoted the clonogenic survival of HCC cells following irradiation compared with empty vector-transfected cells. MUC1 overexpression resulted in >60% reduction in apoptosis induced by irradiation, as determined by Annexin-V/propidium iodide double staining and flow cytometry analysis. Furthermore, overexpression of MUC1 significantly (P<0.05) attenuated the activation of caspase-3 and poly (ADP-ribose) polymerase in response to irradiation exposure. Mechanistically, MUC1 inhibited irradiation-induced apoptosis through activation of janus kinase 2 (JAK2) and STAT3, and induction of anti-apoptotic proteins induced myeloid leukemia cell differentiation protein Mcl-1 (Mcl-1) and BCL2 like 1 (Bcl-xL). Small hairpin RNA-mediated knockdown of STAT3 or MUC1 resensitized MUC1-overexpressing cells to irradiation-induced apoptosis, which was accompanied by reduced expression of Bcl-xL and Mcl-1. Collectively, MUC1 contributes to radioresistance of HCC cells likely through activation of the JAK2/STAT3 signaling pathway and thus represents a potential target for improving radiotherapy against HCC.
Collapse
Affiliation(s)
- Feng-Tao Yi
- Department of Radiotherapy, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Qi-Ping Lu
- Department of General Surgery, Wuhan General Hospital of Guangzhou Command, People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
25
|
Kotzsch M, Kirchner T, Soelch S, Schäfer S, Friedrich K, Baretton G, Magdolen V, Luther T. Inverse association of rab31 and mucin-1 (CA15-3) antigen levels in estrogen receptor-positive (ER+) breast cancer tissues with clinicopathological parameters and patients' prognosis. Am J Cancer Res 2017; 7:1959-1970. [PMID: 28979817 PMCID: PMC5622229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/13/2017] [Indexed: 06/07/2023] Open
Abstract
Dysregulated expression of rab31, a member of the large Rab protein family of the Ras superfamily of small GTPases, has been observed in several types of cancer, including breast cancer. Rab31, depending on its expression level, may regulate the switch between an invasive versus proliferative phenotype of breast cancer cells in vitro. Moreover, gene expression of rab31 is induced by the C-terminal subunit of mucin-1 (MUC1-C) and estrogen receptors (ER). To gain further insights into the clinical relevance of rab31 and mucin-1 expression in breast cancer, we analyzed the relation between rab31 and mucin-1 (CA15-3) antigen levels in detergent tissue extracts of ER-positive (ER+) tumors and clinicopathological parameters as well as patients' prognosis. No significant correlation was observed between rab31 and CA15-3 antigen levels. Elevated rab31 antigen levels in tumor tissue extracts were significantly associated with higher tumor grade (P = 0.021). Strikingly, an inverse significant association was observed for CA15-3 with tumor grade (P = 0.032). Furthermore, high rab31 antigen levels were significantly associated with a high S-phase fraction (SPF, P = 0.047), whereas a trend for lower CA15-3 antigen levels in tumor tissue displaying higher SPF was observed. High rab31 antigen levels were significantly associated with poor 5-year disease-free survival (DFS) of ER+ breast cancer patients in univariate Cox regression analysis (HR = 1.91, 95% CI = 1.14-3.17, P = 0.013). In contrast, high levels of CA15-3 antigen levels were associated with better patients' prognosis (HR = 0.56, 95% CI = 0.33-0.95, P = 0.031). In multivariable analysis, rab31 antigen levels contributed independent prognostic information for DFS when adjusted for prognostically relevant clinicopathological parameters with a HR for high versus low values of 1.97 (95% CI = 1.09-3.54, P = 0.024), whereas CA15-3 antigen levels were not significant. Our results strongly suggest that rab31 antigen levels in tumor tissue are associated with the proliferative status, and rab31 represents an independent biomarker for prognosis in ER+ breast cancer patients. Total mucin-1 (CA 15-3) levels are rather inversely associated with tumor grade and SPF, and elevated levels even indicate prolonged DFS in ER+ breast cancer patients.
Collapse
Affiliation(s)
- Matthias Kotzsch
- Medizinisches Labor OstsachsenD-02526 Bautzen, Germany
- Institut für Pathologie, Technische Universität DresdenD-01307 Dresden, Germany
| | | | - Susanne Soelch
- Klinische Forschergruppe, Klinik für Frauenheilkunde und Geburtshilfe, Technische Universität MünchenD-81675 München, Germany
| | - Sonja Schäfer
- Klinische Forschergruppe, Klinik für Frauenheilkunde und Geburtshilfe, Technische Universität MünchenD-81675 München, Germany
| | - Katrin Friedrich
- Institut für Pathologie, Technische Universität DresdenD-01307 Dresden, Germany
| | - Gustavo Baretton
- Institut für Pathologie, Technische Universität DresdenD-01307 Dresden, Germany
| | - Viktor Magdolen
- Klinische Forschergruppe, Klinik für Frauenheilkunde und Geburtshilfe, Technische Universität MünchenD-81675 München, Germany
| | - Thomas Luther
- Medizinisches Labor OstsachsenD-02526 Bautzen, Germany
- Institut für Pathologie, Technische Universität DresdenD-01307 Dresden, Germany
| |
Collapse
|
26
|
Merikhian P, Ghadirian R, Farahmand L, Mansouri S, Majidzadeh-A K. MUC1 induces tamoxifen resistance in estrogen receptor-positive breast cancer. Expert Rev Anticancer Ther 2017; 17:607-613. [DOI: 10.1080/14737140.2017.1340837] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- Parnaz Merikhian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Reyhane Ghadirian
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Sepideh Mansouri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Phelps CA, Lai SC, Mu D. Roles of Thyroid Transcription Factor 1 in Lung Cancer Biology. VITAMINS AND HORMONES 2017; 106:517-544. [PMID: 29407447 PMCID: PMC11528467 DOI: 10.1016/bs.vh.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Thyroid transcription factor 1 (TTF-1 or NKX2-1) is a transcription factor of fundamental importance in driving lung maturation and morphogenesis. In the last decade, scientists began to appreciate the functional roles of TTF-1 in lung tumorigenesis. This movement was triggered by the discoveries of genetic alterations of TTF-1 in the form of gene amplification in lung cancer. Many downstream target genes of TTF-1 relevant to the lung cancer biology of TTF-1 have been documented. One of the most surprising findings was that TTF-1 may exhibit either pro- or antitumorigenic activities, an outcome with the complexity exceeding the original anticipation purely based on the fact that TTF-1 undergoes gene amplification in lung cancer. In the coming decade, we believe, we will witness additional surprises as the research exploring the cancer roles of TTF-1 progresses.
Collapse
Affiliation(s)
- Cody A Phelps
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Shao-Chiang Lai
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - David Mu
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
28
|
BASP1 interacts with oestrogen receptor α and modifies the tamoxifen response. Cell Death Dis 2017; 8:e2771. [PMID: 28492543 PMCID: PMC5520704 DOI: 10.1038/cddis.2017.179] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/14/2017] [Accepted: 03/15/2017] [Indexed: 01/01/2023]
Abstract
Tamoxifen binds to oestrogen receptor α (ERα) to elicit distinct responses that vary by cell/tissue type and status, but the factors that determine these differential effects are unknown. Here we report that the transcriptional corepressor BASP1 interacts with ERα and in breast cancer cells, this interaction is enhanced by tamoxifen. We find that BASP1 acts as a major selectivity factor in the transcriptional response of breast cancer cells to tamoxifen. In all, 40% of the genes that are regulated by tamoxifen in breast cancer cells are BASP1 dependent, including several genes that are associated with tamoxifen resistance. BASP1 elicits tumour-suppressor activity in breast cancer cells and enhances the antitumourigenic effects of tamoxifen treatment. Moreover, BASP1 is expressed in breast cancer tissue and is associated with increased patient survival. Our data have identified BASP1 as an ERα cofactor that has a central role in the transcriptional and antitumourigenic effects of tamoxifen.
Collapse
|
29
|
Liang YK, Zeng D, Xiao YS, Wu Y, Ouyang YX, Chen M, Li YC, Lin HY, Wei XL, Zhang YQ, Kruyt FAE, Zhang GJ. MCAM/CD146 promotes tamoxifen resistance in breast cancer cells through induction of epithelial-mesenchymal transition, decreased ERα expression and AKT activation. Cancer Lett 2017; 386:65-76. [PMID: 27838413 DOI: 10.1016/j.canlet.2016.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/01/2016] [Accepted: 11/02/2016] [Indexed: 02/05/2023]
Abstract
Tamoxifen resistance presents a prominent clinical challenge in endocrine therapy for hormone sensitive breast cancer. However, the underlying mechanisms that contribute to tamoxifen resistance are not fully understood. In this study, we established a tamoxifen resistant MCF-7 cell line (MCF-7-Tam-R) by continuously incubating MCF-7 cells with 4-OH-tamoxifen. We found that melanoma cell adhesion molecule (MCAM/CD146), a unique epithelial-to-mesenchymal transition (EMT) inducer, was significantly up-regulated at both mRNA and protein levels in MCF-7-Tam-R cells compared to parental MCF-7 cells. Mechanistic research demonstrated that MCAM promotes tamoxifen resistance by transcriptionally suppressing ERα expression and activating the AKT pathway, followed by induction of EMT. Elevated MCAM expression was inversely correlated with recurrence-free and distant metastasis-free survival in a cohort of 4142 patients with breast cancer derived from a public database, particularly in the subgroup only treated with tamoxifen. These results demonstrate a novel function of MCAM in conferring tamoxifen resistance in breast cancer. Targeting MCAM might be a promising therapeutic strategy to overcome tamoxifen resistance in breast cancer patients.
Collapse
Affiliation(s)
- Yuan-Ke Liang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China; Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | - De Zeng
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China; Department of Breast Medical Oncology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China
| | - Ying-Sheng Xiao
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yang Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yan-Xiu Ouyang
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Min Chen
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Yao-Chen Li
- ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Hao-Yu Lin
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China
| | - Yong-Qu Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China
| | - Frank A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Guo-Jun Zhang
- The Breast Center, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou, China; ChangJiang Scholar's Laboratory of Shantou University Medical College, 22 Xinling Road, Shantou, China.
| |
Collapse
|
30
|
Tecalco-Cruz AC, Ramírez-Jarquín JO. Mechanisms that Increase Stability of Estrogen Receptor Alpha in Breast Cancer. Clin Breast Cancer 2016; 17:1-10. [PMID: 27561704 DOI: 10.1016/j.clbc.2016.07.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/29/2016] [Accepted: 07/20/2016] [Indexed: 12/20/2022]
Abstract
Estrogen receptor alpha (ER) is a transcriptional regulator that controls the expression of genes related to cellular proliferation and differentiation in normal mammary tissue. However, the expression, abundance, and activity of this receptor are increased in 70% of breast cancers. The ER upregulation is facilitated by several molecular mechanisms, including protein stability, which represents an important strategy to maintain an active and functional repertoire of ER. Several proteins interact and protect ER from degradation by the ubiquitin-proteasome system. Through diverse mechanisms, these proteins prevent polyubiquitination and degradation of ER, leading to an increase in ER protein levels; consequently, estrogen signaling and its physiologic effects are enhanced in breast cancer cells. Thus, increased protein stability seems to be one of the main reasons that ER is upregulated in breast cancer. Here, we highlight findings on the proteins and mechanisms that participate directly or indirectly in ER stability and their relevance to breast cancer.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F., Mexico.
| | - Josué O Ramírez-Jarquín
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., Mexico
| |
Collapse
|
31
|
Hiraki M, Suzuki Y, Alam M, Hinohara K, Hasegawa M, Jin C, Kharbanda S, Kufe D. MUC1-C Stabilizes MCL-1 in the Oxidative Stress Response of Triple-Negative Breast Cancer Cells to BCL-2 Inhibitors. Sci Rep 2016; 6:26643. [PMID: 27217294 PMCID: PMC4877578 DOI: 10.1038/srep26643] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/06/2016] [Indexed: 01/15/2023] Open
Abstract
Aberrant expression of myeloid cell leukemia-1 (MCL-1) is a major cause of drug resistance in triple-negative breast cancer (TNBC) cells. Mucin 1 (MUC1) is a heterodimeric oncoprotein that is aberrantly overexpressed in most TNBC. The present studies show that targeting the oncogenic MUC1 C-terminal subunit (MUC1-C) in TNBC cells with silencing or pharmacologic inhibition with GO-203 is associated with downregulation of MCL-1 levels. Targeting MUC1-C suppresses the MEK → ERK and PI3K → AKT pathways, and in turn destabilizes MCL-1. The small molecules ABT-737 and ABT-263 target BCL-2, BCL-XL and BCL-w, but not MCL-1. We show that treatment with ABT-737 increases reactive oxygen species and thereby MUC1-C expression. In this way, MUC1-C is upregulated in TNBC cells resistant to ABT-737 or ABT-263. We also demonstrate that MUC1-C is necessary for the resistance-associated increases in MCL-1 levels. Significantly, combining GO-203 with ABT-737 is synergistic in inhibiting survival of parental and drug resistant TNBC cells. These findings indicate that targeting MUC1-C is a potential strategy for reversing MCL-1-mediated resistance in TNBC.
Collapse
Affiliation(s)
- Masayuki Hiraki
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Yozo Suzuki
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Maroof Alam
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Kunihiko Hinohara
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Masanori Hasegawa
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Caining Jin
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Surender Kharbanda
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| | - Donald Kufe
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA 02215, USA
| |
Collapse
|
32
|
Zeng H, Xu W. Ctr9, a key subunit of PAFc, affects global estrogen signaling and drives ERα-positive breast tumorigenesis. Genes Dev 2016; 29:2153-67. [PMID: 26494790 PMCID: PMC4617979 DOI: 10.1101/gad.268722.115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Zeng and Xu discovered that Ctr9, a key subunit of hPAFc, is a central regulator of estrogen signaling that drives ERα+ breast tumorigenesis, rendering it a potential target for the treatment of ERα+ breast cancer. The human RNA polymerase II (RNAPII)-associated factor complex (hPAFc) and its individual subunits have been implicated in human diseases, including cancer. However, its involvement in breast cancer awaits investigation. Using data mining and human breast cancer tissue microarrays, we found that Ctr9, the key scaffold subunit in hPAFc, is highly expressed in estrogen receptor α-positive (ERα+) luminal breast cancer, and the high expression of Ctr9 correlates with poor prognosis. Knockdown of Ctr9 in ERα+ breast cancer cells almost completely erased estrogen-regulated transcriptional response. At the molecular level, Ctr9 enhances ERα protein stability, promotes recruitment of ERα and RNAPII, and stimulates transcription elongation and transcription-coupled histone modifications. Knockdown of Ctr9, but not other hPAFc subunits, alters the morphology, proliferative capacity, and tamoxifen sensitivity of ERα+ breast cancer cells. Together, our study reveals that Ctr9, a key subunit of hPAFc, is a central regulator of estrogen signaling that drives ERα+ breast tumorigenesis, rendering it a potential target for the treatment of ERα+ breast cancer.
Collapse
Affiliation(s)
- Hao Zeng
- McArdle Laboratory for Cancer Research, Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Wei Xu
- McArdle Laboratory for Cancer Research, Graduate Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
33
|
Lakshmanan I, Rachagani S, Hauke R, Krishn SR, Paknikar S, Seshacharyulu P, Karmakar S, Nimmakayala RK, Kaushik G, Johansson SL, Carey GB, Ponnusamy MP, Kaur S, Batra SK, Ganti AK. MUC5AC interactions with integrin β4 enhances the migration of lung cancer cells through FAK signaling. Oncogene 2016; 35:4112-21. [PMID: 26751774 DOI: 10.1038/onc.2015.478] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 10/29/2015] [Accepted: 11/10/2015] [Indexed: 01/07/2023]
Abstract
MUC5AC is a secretory mucin aberrantly expressed in various cancers. In lung cancer, MUC5AC is overexpressed in both primary and metastatic lesions; however, its functional role is not well understood. The present study was aimed at evaluating mechanistic role of MUC5AC on metastasis of lung cancer cells. Clinically, the overexpression of MUC5AC was observed in lung cancer patient tissues and was associated with poor survival. In addition, the overexpression of Muc5ac was also observed in genetically engineered mouse lung adenocarcinoma tissues (Kras(G12D); Trp53(R172H/+); AdCre) in comparison with normal lung tissues. Our functional studies showed that MUC5AC knockdown resulted in significantly decreased migration in two lung cancer cell lines (A549 and H1437) as compared with scramble cells. Expression of integrins (α5, β1, β3, β4 and β5) was decreased in MUC5AC knockdown cells. As both integrins and MUC5AC have a von Willebrand factor domain, we assessed for possible interaction of MUC5AC and integrins in lung cancer cells. MUC5AC strongly interacted only with integrin β4. The co-localization of MUC5AC and integrin β4 was observed both in A549 lung cancer cells as well as genetically engineered mouse adenocarcinoma tissues. Activated integrins recruit focal adhesion kinase (FAK) that mediates metastatic downstream signaling pathways. Phosphorylation of FAK (Y397) was decreased in MUC5AC knockdown cells. MUC5AC/integrin β4/FAK-mediated lung cancer cell migration was confirmed through experiments utilizing a phosphorylation (Y397)-specific FAK inhibitor. In conclusion, overexpression of MUC5AC is a poor prognostic marker in lung cancer. MUC5AC interacts with integrin β4 that mediates phosphorylation of FAK at Y397 leading to lung cancer cell migration.
Collapse
Affiliation(s)
- I Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - R Hauke
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S R Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Paknikar
- Division of Oncology-Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Seshacharyulu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S Karmakar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - R K Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - G Kaushik
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S L Johansson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center University of Nebraska Medical Center, Omaha, NE, USA
| | - G B Carey
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - M P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center University of Nebraska Medical Center, Omaha, NE, USA
| | - S Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - S K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center University of Nebraska Medical Center, Omaha, NE, USA
| | - A K Ganti
- Division of Oncology-Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA.,Department of Internal Medicine, VA Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
34
|
Lloyd SM, Arnold J, Sreekumar A. Metabolomic profiling of hormone-dependent cancers: a bird's eye view. Trends Endocrinol Metab 2015; 26:477-85. [PMID: 26242817 PMCID: PMC4560106 DOI: 10.1016/j.tem.2015.07.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 06/19/2015] [Accepted: 07/01/2015] [Indexed: 01/18/2023]
Abstract
Hormone-dependent cancers present a significant public health challenge, because they are among the most common cancers in the world. One factor associated with cancer development and progression is metabolic reprogramming. By understanding these alterations, we can identify potential markers and novel biochemical therapeutic targets. Metabolic profiling is an advanced technology that allows investigators to assess low-molecular-weight compounds that reflect physiological alterations. Current research in metabolomics on prostate (PCa) and breast cancer (BCa) have made great strides in uncovering specific metabolic pathways that are associated with cancer development, progression, and resistance. In this review, we highlight some of the major findings and potential therapeutic advances that have been reported utilizing this technology.
Collapse
Affiliation(s)
- Stacy M Lloyd
- Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - James Arnold
- Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Arun Sreekumar
- Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
35
|
Hasegawa M, Sinha RK, Kumar M, Alam M, Yin L, Raina D, Kharbanda A, Panchamoorthy G, Gupta D, Singh H, Kharbanda S, Kufe D. Intracellular Targeting of the Oncogenic MUC1-C Protein with a Novel GO-203 Nanoparticle Formulation. Clin Cancer Res 2015; 21:2338-47. [PMID: 25712682 DOI: 10.1158/1078-0432.ccr-14-3000] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/11/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE The MUC1-C oncoprotein is an intracellular target that is druggable with cell-penetrating peptide inhibitors. However, development of peptidyl drugs for treating cancer has been a challenge because of unfavorable pharmacokinetic parameters and limited cell-penetrating capabilities. EXPERIMENTAL DESIGN Encapsulation of the MUC1-C inhibitor GO-203 in novel polymeric nanoparticles was studied for effects on intracellular targeting of MUC1-C signaling and function. RESULTS Our results show that loading GO-203 into tetrablock polylactic acid (PLA)-polyethylene glycol (PEG)-polypropylene glycol (PPG)-PEG copolymers is achievable and, notably, is enhanced by increasing PEG chain length. In addition, we found that release of GO-203 from these nanoparticles is controllable over at least 7 days. GO-203/nanoparticle treatment of MUC1-C-positive breast and lung cancer cells in vitro was more active with less frequent dosing than that achieved with nonencapsulated GO-203. Moreover, treatment with GO-203/nanoparticles blocked MUC1-C homodimerization, consistent with on-target effects. GO-203/nanoparticle treatment was also effective in downregulating TIGAR, disrupting redox balance, and inhibiting the self-renewal capacity of cancer cells. Significantly, weekly administration of GO-203/nanoparticles to mice bearing syngeneic or xenograft tumors was associated with regressions that were comparable with those found when dosing on a daily basis with GO-203. CONCLUSIONS These findings thus define an effective approach for (i) sustained administration of GO-203 in polymeric PLA-(PEG-PPG-PEG) nanoparticles to target MUC1-C in cancer cells and (ii) the potential delivery of other anticancer peptide drugs.
Collapse
Affiliation(s)
- Masanori Hasegawa
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Raj Kumar Sinha
- Center for Biomedical Engineering, Indian Institute of Technology, New Delhi, India
| | - Manoj Kumar
- Center for Biomedical Engineering, Indian Institute of Technology, New Delhi, India
| | - Maroof Alam
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Li Yin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Akriti Kharbanda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | | | - Dikshi Gupta
- Center for Biomedical Engineering, Indian Institute of Technology, New Delhi, India
| | - Harpal Singh
- Center for Biomedical Engineering, Indian Institute of Technology, New Delhi, India.
| | | | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
36
|
Alam M, Rajabi H, Ahmad R, Jin C, Kufe D. Targeting the MUC1-C oncoprotein inhibits self-renewal capacity of breast cancer cells. Oncotarget 2015; 5:2622-34. [PMID: 24770886 PMCID: PMC4058032 DOI: 10.18632/oncotarget.1848] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The capacity of breast cancer cells to form mammospheres in non-adherent serum-free culture is used as a functional characteristic of the self-renewing stem-like cell population. The present studies demonstrate that silencing expression of the MUC1-C oncoprotein inhibits growth of luminal MCF-7 and HER2-overexpressing SKBR3 breast cancer cells as mammospheres. We also show that triple-negative MDA-MB-468 breast cancer cells are dependent on MUC1-C for growth as mammospheres and tumor xenografts. Similar results were obtained when MUC1-C function was inhibited by expression of a MUC1-C(CQC→AQA) mutant. Moreover, treatment with the MUC1-C inhibitor GO-203, a cell penetrating peptide that binds to the MUC1-C cytoplasmic domain and blocks MUC1-C function, confirmed the importance of this target for self-renewal. The mechanistic basis for these findings is supported by the demonstration that MUC1-C activates NF-κB, occupies the IL-8 promoter with NF-κB, and induces IL-8 transcription. MUC1-C also induces NF-κB-dependent expression of the IL-8 receptor, CXCR1. In concert with these results, targeting MUC1-C with GO-203 suppresses IL-8/CXCR1 expression and disrupts the formation of established mammospheres. Our findings indicate that MUC1-C contributes to the self-renewal of breast cancer cells by activating the NF-κB→IL-8/CXCR1 pathway and that targeting MUC1-C represents a potential approach for the treatment of this population.
Collapse
Affiliation(s)
- Maroof Alam
- Dana-Farber Cancer Institute Harvard Medical School Boston, MA
| | | | | | | | | |
Collapse
|
37
|
Bouillez A, Gnemmi V, Gaudelot K, Hémon B, Ringot B, Pottier N, Glowacki F, Butruille C, Cauffiez C, Hamdane M, Sergeant N, Van Seuningen I, Leroy X, Aubert S, Perrais M. MUC1-C nuclear localization drives invasiveness of renal cancer cells through a sheddase/gamma secretase dependent pathway. Oncotarget 2015; 5:754-63. [PMID: 24504508 PMCID: PMC3996672 DOI: 10.18632/oncotarget.1768] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MUC1 is a membrane-anchored mucin and its cytoplasmic tail (CT) can interact with many signaling pathways and act as a co-transcription factor to activate genes involved in tumor progression and metastasis. MUC1 is overexpressed in renal cell carcinoma with correlation to prognosis and has been implicated in the hypoxic pathway, the main renal carcinogenetic pathway. In this context, we assessed the effects of MUC1 overexpression on renal cancer cells properties. Using shRNA strategy and/or different MUC1 constructs, we found that MUC1-extracellular domain and MUC1-CT are involved in increase of migration, cell viability, resistance to anoikis and in decrease of cell aggregation in cancer cells. Invasiveness depends only on MUC1-CT. Then, by using siRNA strategy and/or pharmacological inhibitors or peptides, we showed that sheddases ADAM10, ADAM17 and gamma-secretase are necessary for MUC1 C-terminal subunit (MUC1-C) nuclear location and in increase of invasion property. Finally, MUC1 overexpression increases ADAM10/17 protein expression suggesting a positive regulatory loop. In conclusion, we report that MUC1 acts in renal cancer progression and MUC1-C nuclear localization drives invasiveness of cancer cells through a sheddase/gamma secretase dependent pathway. MUC1 appears as a therapeutic target by blocking MUC1 cleavage or nuclear translocation by using pharmacological approach and peptide strategies.
Collapse
Affiliation(s)
- Audrey Bouillez
- Inserm, UMR837, Equipe 5 "Mucines, différenciation et cancérogenèse épithéliales", Jean-Pierre Aubert Research Center, Lille Cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Background The majority of genetic biomarkers for human cancers are defined by statistical screening of high-throughput genomics data. While a large number of genetic biomarkers have been proposed for diagnostic and prognostic applications, only a small number have been applied in the clinic. Similarly, the use of proteomics methods for the discovery of cancer biomarkers is increasing. The emerging field of proteogenomics seeks to enrich the value of genomics and proteomics approaches by studying the intersection of genomics and proteomics data. This task is challenging due to the complex nature of transcriptional and translation regulatory mechanisms and the disparities between genomic and proteomic data from the same samples. In this study, we have examined tumor antigens as potential biomarkers for breast cancer using genomics and proteomics data from previously reported laser capture microdissected ER+ tumor samples. Results We applied proteogenomic analyses to study the genetic aberrations of 32 tumor antigens determined in the proteomic data. We found that tumor antigens that are aberrantly expressed at the genetic level and expressed at the protein level, are likely involved in perturbing pathways directly linked to the hallmarks of cancer. The results found by proteogenomic analysis of the 32 tumor antigens studied here, capture largely the same pathway irregularities as those elucidated from large-scale screening of genomics analyses, where several thousands of genes are often found to be perturbed. Conclusion Tumor antigens are a group of proteins recognized by the cells of the immune system. Specifically, they are recognized in tumor cells where they are present in larger than usual amounts, or are physiochemically altered to a degree at which they no longer resemble native human proteins. This proteogenomic analysis of 32 tumor antigens suggests that tumor antigens have the potential to be highly specific biomarkers for different cancers.
Collapse
|
39
|
Alam M, Ahmad R, Rajabi H, Kufe D. MUC1-C Induces the LIN28B→LET-7→HMGA2 Axis to Regulate Self-Renewal in NSCLC. Mol Cancer Res 2014; 13:449-60. [PMID: 25368430 DOI: 10.1158/1541-7786.mcr-14-0363] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
UNLABELLED The LIN28B→let-7 pathway contributes to regulation of the epithelial-mesenchymal transition (EMT) and stem cell self-renewal. The oncogenic MUC1-C transmembrane protein is aberrantly overexpressed in lung and other carcinomas; however, there is no known association between MUC1-C and the LIN28B→let-7 pathway. Here in non-small cell lung cancer (NSCLC), silencing MUC1-C downregulates the RNA-binding protein LIN28B and coordinately increases the miRNA let-7. Targeting MUC1-C function with a dominant-negative mutant or a peptide inhibitor provided confirming evidence that MUC1-C induces LIN28B→let-7 signaling. Mechanistically, MUC1-C promotes NF-κB p65 chromatin occupancy of the LIN28B first intron and activates LIN28B transcription, which is associated with suppression of let-7. Consistent with let-7-mediated inhibition of HMGA2 transcripts, targeting of MUC1-C also decreases HMGA2 expression. HMGA2 has been linked to stemness, and functions as a competing endogenous RNA (ceRNA) of let-7-mediated regulation of the TGFβ coreceptor TGFBR3. Accordingly, targeting MUC1-C suppresses HMGA2 mRNA and protein, which is associated with decreases in TGFBR3, reversal of the EMT phenotype, and inhibition of self-renewal capacity. These findings support a model in which MUC1-C activates the ⇑LIN28B→⇓let-7→⇑HMGA2 axis in NSCLC and thereby promotes EMT traits and stemness. IMPLICATIONS A novel pathway is defined in which MUC1-C drives LIN28B→let-7→HMGA2 signaling, EMT, and self-renewal in NSCLC.
Collapse
Affiliation(s)
- Maroof Alam
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rehan Ahmad
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hasan Rajabi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Donald Kufe
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
40
|
Jonckheere N, Skrypek N, Van Seuningen I. Mucins and tumor resistance to chemotherapeutic drugs. Biochim Biophys Acta Rev Cancer 2014; 1846:142-51. [PMID: 24785432 DOI: 10.1016/j.bbcan.2014.04.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Revised: 04/22/2014] [Accepted: 04/23/2014] [Indexed: 12/30/2022]
Abstract
Epithelial cancer patients not considered eligible for surgical resection frequently benefit from chemotherapy. Chemotherapy is the treatment of cancer with one or combination of cytotoxic or cytostatic drugs. Recent advances in chemotherapy allowed a great number of cancer patients to receive treatment with significant results. Unfortunately, resistance to chemotherapeutic drug treatment is a major challenge for clinicians in the majority of epithelial cancers because it is responsible for the inefficiency of therapies. Mucins belong to a heterogeneous group of large O-glycoproteins that can be either secreted or membrane-bound. Implications of mucins have been described in relation to cancer cell behavior and cell signaling pathways associated with epithelial tumorigenesis. Because of the frequent alteration of the pattern of mucin expression in cancers as well as their structural and functional characteristics, mucins are thought to also be involved in response to therapies. In this report, we review the roles of mucins in chemoresistance and the associated underlying molecular mechanisms (physical barrier, resistance to apoptosis, drug metabolism, cell stemness, epithelial-mesenchymal transition) and discuss the therapeutic tools/strategies and/or prognosis biomarkers for personalized chemotherapy that could be proposed from these studies.
Collapse
Affiliation(s)
- Nicolas Jonckheere
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France.
| | - Nicolas Skrypek
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France
| | - Isabelle Van Seuningen
- Inserm, UMR837, Jean Pierre Aubert Research Center, Team #5 "Mucins, Epithelial Differentiation and Carcinogenesis", rue Polonovski, 59045 Lille Cedex, France; Université Lille Nord de France, Lille, France; Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille Cedex, France
| |
Collapse
|
41
|
Schlom J, Hodge JW, Palena C, Tsang KY, Jochems C, Greiner JW, Farsaci B, Madan RA, Heery CR, Gulley JL. Therapeutic cancer vaccines. Adv Cancer Res 2014; 121:67-124. [PMID: 24889529 PMCID: PMC6324585 DOI: 10.1016/b978-0-12-800249-0.00002-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Therapeutic cancer vaccines have the potential of being integrated in the therapy of numerous cancer types and stages. The wide spectrum of vaccine platforms and vaccine targets is reviewed along with the potential for development of vaccines to target cancer cell "stemness," the epithelial-to-mesenchymal transition (EMT) phenotype, and drug-resistant populations. Preclinical and recent clinical studies are now revealing how vaccines can optimally be used with other immune-based therapies such as checkpoint inhibitors, and so-called nonimmune-based therapeutics, radiation, hormonal therapy, and certain small molecule targeted therapies; it is now being revealed that many of these traditional therapies can lyse tumor cells in a manner as to further potentiate the host immune response, alter the phenotype of nonlysed tumor cells to render them more susceptible to T-cell lysis, and/or shift the balance of effector:regulatory cells in a manner to enhance vaccine efficacy. The importance of the tumor microenvironment, the appropriate patient population, and clinical trial endpoints is also discussed in the context of optimizing patient benefit from vaccine-mediated therapy.
Collapse
Affiliation(s)
- Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kwong-Yok Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Caroline Jochems
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - John W Greiner
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Benedetto Farsaci
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ravi A Madan
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - James L Gulley
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
42
|
Jochems C, Tucker JA, Vergati M, Boyerinas B, Gulley JL, Schlom J, Tsang KY. Identification and characterization of agonist epitopes of the MUC1-C oncoprotein. Cancer Immunol Immunother 2013; 63:161-74. [PMID: 24233342 DOI: 10.1007/s00262-013-1494-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 10/19/2013] [Indexed: 12/31/2022]
Abstract
The MUC1 tumor-associated antigen is overexpressed in the majority of human carcinomas and several hematologic malignancies. Much attention has been paid to the hypoglycosylated variable number of tandem repeats (VNTR) region of the N-terminus of MUC1 as a vaccine target, and recombinant viral vector vaccines are also being evaluated that express the entire MUC1 transgene. While previous studies have described MUC1 as a tumor-associated tissue differentiation antigen, studies have now determined that the C-terminus of MUC1 (MUC1-C) is an oncoprotein, and its expression is an indication of poor prognosis in numerous tumor types. We report here the identification of nine potential CD8⁺ cytotoxic T lymphocyte epitopes of MUC1, seven in the C-terminus and two in the VNTR region, and have identified enhancer agonist peptides for each of these epitopes. These epitopes span HLA-A2, HLA-A3, and HLA-A24 major histocompatibility complex (MHC) class I alleles, which encompass the majority of the population. The agonist peptides, compared to the native peptides, more efficiently (a) generate T-cell lines from the peripheral blood mononuclear cells of cancer patients, (b) enhance the production of IFN-γ by peptide-activated human T cells, and (c) lyse human tumor cell targets in an MHC-restricted manner. The agonist epitopes described here can be incorporated into various vaccine platforms and for the ex vivo generation of human T cells. These studies provide the rationale for the T-cell-mediated targeting of the oncogenic MUC1-C, which has been shown to be an important factor in both drug resistance and poor prognosis for numerous tumor types.
Collapse
|