1
|
Bhutta ZA, Choi KC. Phytochemicals as Novel Therapeutics for Triple-Negative Breast Cancer: A Comprehensive Review of Current Knowledge. Phytother Res 2025; 39:364-396. [PMID: 39533509 DOI: 10.1002/ptr.8376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Triple-negative breast cancer is a characteristic subtype of breast cancer that lacks the estrogen receptor, human epidermal growth factor receptor 2, and progesterone receptor. Because of its highly diverse subtypes, increased metastasis capability, and poor prognosis, the risk of mortality for people with triple-negative breast cancers is high as compared with other cancers. Chemotherapy is currently playing a major role in treating triple-negative breast cancer patients; however, poor prognosis due to drug resistance is causing serious concern. Recent studies on several phytochemicals derived from various plants being used in Traditional Chinese Medicine, Traditional Korean Medicine, Ayurveda (Traditional Indian Medicine), and so on, have demonstrated to be a promising agent as a viable therapy against triple-negative breast cancer. Phytochemicals categorized as alkaloids, polyphenols, terpenoids, phytosterols, and organosulfur compounds have been demonstrated to reduce cancer cell proliferation and metastasis by activating various molecular pathways, thereby reducing the spread of triple-negative breast cancer. This review analyzes the molecular mechanisms by which various phytochemicals fight triple-negative breast cancer and offers a perspective on the difficulties and potential prospects for treating triple-negative breast cancer with various phytochemicals.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
2
|
Sinha S, Guo R, Del Busso MD, Han W, Boysen J, Wellik LE, Ghosh AK, Kay NE. Aberrant activation of AXL may drive progression of squamous cell carcinoma in CLL patients: a mechanistic study with clinical implications. Br J Cancer 2024; 131:589-600. [PMID: 38886556 PMCID: PMC11300914 DOI: 10.1038/s41416-024-02752-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Occurrence of squamous cell carcinoma (SCC) even in early-stage, untreated chronic lymphocytic leukemia (CLL) patients can be a significant morbidity issue with occasional transformation into metastatic skin lesions. METHODS CLL cells and extracellular vesicles (EVs) from CLL patients' blood/plasma were purified and used. Expression/activation of AXL and its functions in normal keratinocytes (HEKa) were assessed in vitro co-culture system and in SCC tissues. RESULTS We detected aberrant activation of AXL, AKT and ERK-1/2 in SCC cell lines compared to HEKa. We also detected increased expression of AXL in primary SCC tissues obtained from CLL patients. Increased activation of AXL, AKT, ERK-1/2 and Src was discernible in HEKa upon co-culturing with CLL cells. Further analysis suggests that Gas6, a ligand of AXL, regulates AXL activation in co-cultured HEKa. Interestingly, exposure of HEKa cells to CLL plasma-derived EVs induced expression of AXL, P-AKT, and EMT-associated markers leading to migration of the cells. Finally, pharmacologic inhibition of AXL induced cell death in SCC lines in a dose dependent manner. CONCLUSIONS Our findings that CLL cells likely are involved in driving SCC progression, at least in part, via activation of the AXL signaling axis, indicating that AXL inhibition may be beneficial for our CLL patients with SCC.
Collapse
Affiliation(s)
- Sutapa Sinha
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
| | - Ray Guo
- Division of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Weiguo Han
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Justin Boysen
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Asish K Ghosh
- Stephenson Cancer Center and Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Neil E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
3
|
Shirley CA, Chhabra G, Amiri D, Chang H, Ahmad N. Immune escape and metastasis mechanisms in melanoma: breaking down the dichotomy. Front Immunol 2024; 15:1336023. [PMID: 38426087 PMCID: PMC10902921 DOI: 10.3389/fimmu.2024.1336023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Melanoma is one of the most lethal neoplasms of the skin. Despite the revolutionary introduction of immune checkpoint inhibitors, metastatic spread, and recurrence remain critical problems in resistant cases. Melanoma employs a multitude of mechanisms to subvert the immune system and successfully metastasize to distant organs. Concerningly, recent research also shows that tumor cells can disseminate early during melanoma progression and enter dormant states, eventually leading to metastases at a future time. Immune escape and metastasis have previously been viewed as separate phenomena; however, accumulating evidence is breaking down this dichotomy. Recent research into the progressive mechanisms of melanoma provides evidence that dedifferentiation similar to classical epithelial to mesenchymal transition (EMT), genes involved in neural crest stem cell maintenance, and hypoxia/acidosis, are important factors simultaneously involved in immune escape and metastasis. The likeness between EMT and early dissemination, and differences, also become apparent in these contexts. Detailed knowledge of the mechanisms behind "dual drivers" simultaneously promoting metastatically inclined and immunosuppressive environments can yield novel strategies effective in disabling multiple facets of melanoma progression. Furthermore, understanding progression through these drivers may provide insight towards novel treatments capable of preventing recurrence arising from dormant dissemination or improving immunotherapy outcomes.
Collapse
Affiliation(s)
- Carl A. Shirley
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Deeba Amiri
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
| | - Hao Chang
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin, Madison, WI, United States
- William S. Middleton Memorial Veterans Hospital, Madison, WI, United States
| |
Collapse
|
4
|
Tang Y, Zang H, Wen Q, Fan S. AXL in cancer: a modulator of drug resistance and therapeutic target. J Exp Clin Cancer Res 2023; 42:148. [PMID: 37328828 DOI: 10.1186/s13046-023-02726-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
AXL is a member of the TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases family (RTKs), and its abnormal expression has been linked to clinicopathological features and poor prognosis of cancer patients. There is mounting evidence supporting AXL's role in the occurrence and progression of cancer, as well as drug resistance and treatment tolerance. Recent studies revealed that reducing AXL expression can weaken cancer cells' drug resistance, indicating that AXL may be a promising target for anti-cancer drug treatment. This review aims to summarize the AXL's structure, the mechanisms regulating and activating it, and its expression pattern, especially in drug-resistant cancers. Additionally, we will discuss the diverse functions of AXL in mediating cancer drug resistance and the potential of AXL inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Yaoxiang Tang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
5
|
Brown BA, Lazzara MJ. Single-cell RNA sequencing reveals microenvironment context-specific routes for epithelial-mesenchymal transition in pancreas cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542969. [PMID: 37398348 PMCID: PMC10312528 DOI: 10.1101/2023.05.30.542969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
In the PDAC tumor microenvironment, multiple factors initiate the epithelial-mesenchymal transition (EMT) that occurs heterogeneously among transformed ductal cells, but it is unclear if different drivers promote EMT through common or distinct signaling pathways. Here, we use single-cell RNA sequencing (scRNA-seq) to identify the transcriptional basis for EMT in pancreas cancer cells in response to hypoxia or EMT-inducing growth factors. Using clustering and gene set enrichment analysis, we find EMT gene expression patterns that are unique to the hypoxia or growth factor conditions or that are common between them. Among the inferences from the analysis, we find that the FAT1 cell adhesion protein is enriched in epithelial cells and suppresses EMT. Further, the receptor tyrosine kinase AXL is preferentially expressed in hypoxic mesenchymal cells in a manner correlating with YAP nuclear localization, which is suppressed by FAT1 expression. AXL inhibition prevents EMT in response to hypoxia but not growth factors. Relationships between FAT1 or AXL expression with EMT were confirmed through analysis of patient tumor scRNA-seq data. Further exploration of inferences from this unique dataset will reveal additional microenvironment context-specific signaling pathways for EMT that may represent novel drug targets for PDAC combination therapies.
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW The AXL signaling pathway is associated with tumor growth as well as poor prognosis in cancer. Here, we highlight recent strategies for targeting AXL in the treatment of solid and hematological malignancies. RECENT FINDINGS AXL is a key player in survival, metastasis, and therapeutic resistance in many cancers. A range of AXL-targeted therapies, including tyrosine kinase inhibitors, monoclonal antibodies, antibody-drug conjugates, and soluble receptors, have entered clinical development. Notably, AXL inhibitors in combination with immune checkpoint inhibitors demonstrate early promise; however, further understanding of predictive biomarkers and treatment sequencing is necessary. Based on its role in tumor growth and drug resistance, AXL represents a promising therapeutic target in oncology. Results from ongoing clinical trials will provide valuable insights into the role of AXL inhibitors, both as single agents and in combination with other therapies.
Collapse
Affiliation(s)
- Sheena Bhalla
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
- Division of Hematology-Oncology, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - David E Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), UT Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
- Peter O'Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
7
|
Harnessing epithelial-mesenchymal plasticity to boost cancer immunotherapy. Cell Mol Immunol 2023; 20:318-340. [PMID: 36823234 PMCID: PMC10066239 DOI: 10.1038/s41423-023-00980-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/17/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy is a powerful option for cancer treatment. Despite demonstrable progress, most patients fail to respond or achieve durable responses due to primary or acquired ICB resistance. Recently, tumor epithelial-to-mesenchymal plasticity (EMP) was identified as a critical determinant in regulating immune escape and immunotherapy resistance in cancer. In this review, we summarize the emerging role of tumor EMP in ICB resistance and the tumor-intrinsic or extrinsic mechanisms by which tumors exploit EMP to achieve immunosuppression and immune escape. We discuss strategies to modulate tumor EMP to alleviate immune resistance and to enhance the efficiency of ICB therapy. Our discussion provides new prospects to enhance the ICB response for therapeutic gain in cancer patients.
Collapse
|
8
|
Farghadani R, Naidu R. Curcumin: Modulator of Key Molecular Signaling Pathways in Hormone-Independent Breast Cancer. Cancers (Basel) 2021; 13:cancers13143427. [PMID: 34298639 PMCID: PMC8307022 DOI: 10.3390/cancers13143427] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Breast cancer remains the most commonly diagnosed cancer and the leading cause of cancer death among females worldwide. It is a highly heterogeneous disease, classified according to hormone and growth factor receptor expression. Patients with triple negative breast cancer (TNBC) (estrogen receptor-negative/progesterone receptor-negative/human epidermal growth factor receptor (HER2)-negative) and hormone-independent HER2 overexpressing subtypes still represent highly aggressive behavior, metastasis, poor prognosis, and drug resistance. Thus, new alternative anticancer agents based on the use of natural products have been receiving enormous attention. In this regard, curcumin is a promising lead in cancer drug discovery due its ability to modulate a diverse range of molecular targets and signaling pathways. The current review has emphasized the underlying mechanism of curcumin anticancer action mediated through the modulation of PI3K/Akt/mTOR, JAK/STAT, MAPK, NF-ĸB, p53, Wnt/β-catenin, apoptosis, and cell cycle pathways in hormone-independent breast cancer, providing frameworks for future studies and insights to improve its efficiency in clinical practice. Abstract Breast cancer is the most frequently diagnosed cancer and the leading cause of cancer death among women worldwide. Despite the overall successes in breast cancer therapy, hormone-independent HER2 negative breast cancer, also known as triple negative breast cancer (TNBC), lacking estrogens and progesterone receptors and with an excessive expression of human epidermal growth factor receptor 2 (HER2), along with the hormone-independent HER2 positive subtype, still remain major challenges in breast cancer treatment. Due to their poor prognoses, aggressive phenotype, and highly metastasis features, new alternative therapies have become an urgent clinical need. One of the most noteworthy phytochemicals, curcumin, has attracted enormous attention as a promising drug candidate in breast cancer prevention and treatment due to its multi-targeting effect. Curcumin interrupts major stages of tumorigenesis including cell proliferation, survival, angiogenesis, and metastasis in hormone-independent breast cancer through the modulation of multiple signaling pathways. The current review has highlighted the anticancer activity of curcumin in hormone-independent breast cancer via focusing on its impact on key signaling pathways including the PI3K/Akt/mTOR pathway, JAK/STAT pathway, MAPK pathway, NF-ĸB pathway, p53 pathway, and Wnt/β-catenin, as well as apoptotic and cell cycle pathways. Besides, its therapeutic implications in clinical trials are here presented.
Collapse
|
9
|
Han S, Wang Y, Ge C, Gao M, Wang X, Wang F, Sun L, Li S, Dong T, Dang Z, Cui W, Zhang G, Liu N. Pharmaceutical inhibition of AXL suppresses tumor growth and invasion of esophageal squamous cell carcinoma. Exp Ther Med 2020; 20:41. [PMID: 32952632 PMCID: PMC7480165 DOI: 10.3892/etm.2020.9169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/01/2020] [Indexed: 01/02/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common type of cancer in a number of regions of the world, including East Asia, South Africa and Iran. It is often associated with poor prognosis rates. Tyrosine-protein kinase receptor UFO (AXL) is overexpressed in a subset of ESCC tumors, therefore the present study aimed to determine the effect of R428, a selective inhibitor of AXL, on ESCC tumor cells. TE1 and KYSE150 cell lines were used as models to investigate the effects of R428 treatment. The proliferative rate of the tumor cells was analyzed using MTT and colony formation assays. In addition, cell migration and invasion rates were analyzed using wound healing and Matrigel assays, respectively. The expression levels of matrix metalloproteinase (MMP)2 and MMP9, and the activation of protein kinase B (AKT), extracellular signal-regulated kinase (ERK) and AXL signaling were analyzed using gelatin zymography and western blotting. The results revealed that R428 inhibited the proliferative and invasive abilities of both cell lines. Furthermore, AXL, AKT and ERK signaling were all decreased in response to R428 treatment, alongside the expression levels of MMP2 and MMP9. In conclusion, the results of the present study suggested that R428 treatment may suppress ESCC tumor cell proliferation and invasion, representing a potential therapeutic target for ESCC.
Collapse
Affiliation(s)
- Sha Han
- Collaborative Innovation Center for Birth Defect Research and Transformation of Shandong Province, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Yequan Wang
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Chengyan Ge
- The Second Medical College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Mingtao Gao
- The Second Medical College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Xintong Wang
- The Second Medical College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Feiyu Wang
- The Second Medical College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Lei Sun
- The Second Medical College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Sheng Li
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Tingting Dong
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Zhen Dang
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Wen Cui
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Guoan Zhang
- Institute of Forensic Medicine and Laboratory Medicine, Forensic Science Center of Jining, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Ning Liu
- Information Technology Centre, Jining Medical University, Jining, Shandong 272067, P.R. China
| |
Collapse
|
10
|
Kong L, Lu X, Chen X, Wu Y, Zhang Y, Shi H, Li J. Qigesan inhibits esophageal cancer cell invasion and migration by inhibiting Gas6/Axl-induced epithelial-mesenchymal transition. Aging (Albany NY) 2020; 12:9714-9725. [PMID: 32432570 PMCID: PMC7288918 DOI: 10.18632/aging.103238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/31/2020] [Indexed: 12/14/2022]
Abstract
Qigesan (QGS) has been used to effectively treat esophageal cancer (EC) for decades in China, but the mechanism by which it suppresses EC metastasis remains unknown. In this study, we examined the effects of QGS on EC cell mobility. Using immunohistochemistry and immunofluorescence, expression of Gas6 and Axl, which promote tumor cell migration and invasion, was examined in carcinoma tissues and adjacent normal tissues from EC patients. Levels of Gas6, Axl, and the Gas6/Axl complex were also examined in ECA109 and TE13 EC cells treated with QGS. In addition, immunofluorescent staining and quantitative protein analysis were used to examine E-cadherin, N-cadherin, and Snail levels in ECA109 and TE13 EC cells after QSG administration, and cell mobility was assessed. The results demonstrated that levels of Gas6 and Axl expression are higher in EC tissues than in adjacent normal tissues. Moreover, QGS decreased Gas6/Axl levels, increased E-cadherin expression, decreased Snail and N-cadherin expression, and inhibited epithelial-mesenchymal transition (EMT) in EC cells. QGS thus suppresses EMT in EC by inhibiting Gas6/Axl binding.
Collapse
Affiliation(s)
- Lingyu Kong
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Xin Lu
- Department of Clinical Laboratory, Tangshan Maternal and Children Hospital, Tangshan 063000, Hebei, China
| | - Xuanyu Chen
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Yunyan Wu
- Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Yushuang Zhang
- Department of Traditional Chinese Medicine, Tumor Hospital of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Huijuan Shi
- Department of Traditional Chinese Medicine, Tumor Hospital of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, Hebei, China,Department of Traditional Chinese Medicine, Tumor Hospital of Hebei Province, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei, China
| |
Collapse
|
11
|
Yang PW, Liu YC, Chang YH, Lin CC, Huang PM, Hua KT, Lee JM, Hsieh MS. Cabozantinib (XL184) and R428 (BGB324) Inhibit the Growth of Esophageal Squamous Cell Carcinoma (ESCC). Front Oncol 2019; 9:1138. [PMID: 31781483 PMCID: PMC6851194 DOI: 10.3389/fonc.2019.01138] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 10/11/2019] [Indexed: 12/18/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly disease for which no effective targeted therapeutic agent has been approved. Both AXL and c-MET have been reported to be independent prognostic factors for ESCC. Thus, inhibitors of AXL/c-MET might have great potential as targeted therapy for ESCC. In the current study, we evaluated the therapeutic potential of the AXL/c-MET selective inhibitors, R428 and cabozantinib, in cell and mouse xenograft models. We demonstrated that both R428 and cabozantinib significantly inhibited the growth of CE81T and KYSE-70 ESCC cells and showed by wound-healing assay that they both inhibited ESCC cell migration. In the animal model, ESCC xenograft models were established by injecting KYSE-70 cells with Matrigel into the upper back region of NOD-SCID male mice followed by treatment with vehicle control, R428 (50 mg/kg/day), cisplatin (1.0 mg/kg), or cabozantinib (30 mg/kg/day) for the indicated number of days. R428 alone significantly inhibited ESCC tumor growth compared to the vehicle; however, no synergistic effect with cisplatin was observed. Notably, the dramatic efficacy of cabozantinib alone was observed in the mouse xenograft model. Collectively, our study demonstrated that both cabozantinib and R428 inhibit ESCC growth in cell and xenograft models. The results reveal the great potential of using cabozantinib for targeted therapy of ESCC.
Collapse
Affiliation(s)
- Pei-Wen Yang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Cheng Liu
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ya-Han Chang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Ching Lin
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Pei-Ming Huang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Kuo-Tai Hua
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Jang-Ming Lee
- Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Min-Shu Hsieh
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|