1
|
Ferreira T, Azevedo T, Silva J, Faustino-Rocha AI, Oliveira PA. Current views on in vivo models for breast cancer research and related drug development. Expert Opin Drug Discov 2024; 19:189-207. [PMID: 38095187 DOI: 10.1080/17460441.2023.2293152] [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: 07/10/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Animal models play a crucial role in breast cancer research, in particular mice and rats, who develop mammary tumors that closely resemble their human counterparts. These models allow the study of mechanisms behind breast carcinogenesis, as well as the efficacy and safety of new, and potentially more effective and advantageous therapeutic approaches. Understanding the advantages and disadvantages of each model is crucial to select the most appropriate one for the research purpose. AREA COVERED This review provides a concise overview of the animal models available for breast cancer research, discussing the advantages and disadvantages of each one for searching new and more effective approaches to treatments for this type of cancer. EXPERT OPINION Rodent models provide valuable information on the genetic alterations of the disease, the tumor microenvironment, and allow the evaluation of the efficacy of chemotherapeutic agents. However, in vivo models have limitations, and one of them is the fact that they do not fully mimic human diseases. Choosing the most suitable model for the study purpose is crucial for the development of new therapeutic agents that provide better care for breast cancer patients.
Collapse
Affiliation(s)
- Tiago Ferreira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Tiago Azevedo
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Jessica Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Department of Zootechnics, School of Sciences and Technology, University of Évora, Évora, Portugal
- Department of Zootechnics, School of Sciences and Technology, Comprehensive Health Research Center, Évora, Portugal
| | - Paula A Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
- Clinical Academic Center of Trás-Os-Montes and Alto Douro, University of Trás-Os-Montes and Alto Douro, Vila Real, Portugal
| |
Collapse
|
2
|
Ouyang C, Zhang J, Lei X, Xie Z, Liu X, Li Y, Huang S, Wang Z, Tang G. Advances in antitumor research of HIF-1α inhibitor YC-1 and its derivatives. Bioorg Chem 2023; 133:106400. [PMID: 36739684 DOI: 10.1016/j.bioorg.2023.106400] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 01/03/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023]
Abstract
Generally, hypoxia-inducible factor-1α (HIF-1α) is highly expressed in solid tumors, it plays a key role in the occurrence and development of tumors, hindering cancer treatment in various ways. The antitumor activity and pharmacological mechanism of YC-1 [3-(5'-hydroxymethyl-2'-furyl)-1‑benzyl indazole], an HIF-1α inhibitor, and the design and synthesis of its derivatives have attracted tremendous attention in the field of antitumor research. YC-1 is a potential drug candidate and a lead compound for tumor therapy. Hence, the multifaceted mechanism of action of YC-1 and the structure activity relationship (SAR) of its derivatives are important factors to be considered for the development of HIF-1α inhibitors. Therefore, this review aimed to provide a comprehensive overview of the various antitumor mechanisms of YC-1 in antitumor research and an in-depth summary of the SAR for the development of its derivatives. A full understanding and discussion of these aspects are expected to provide potential ideas for developing novel HIF-1α inhibitors and antitumor drugs belonging to the YC-1 class. The review also highlighted the application prospects of the YC-1 class of potential antitumor candidates, and provided some unique insights about these antitumor agents.
Collapse
Affiliation(s)
- Chenglin Ouyang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medicial School, University of South China, Hengyang, Hunan 421001, China
| | - Jing Zhang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medicial School, University of South China, Hengyang, Hunan 421001, China
| | - Xiaoyong Lei
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medicial School, University of South China, Hengyang, Hunan 421001, China
| | - Zhizhong Xie
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medicial School, University of South China, Hengyang, Hunan 421001, China
| | - Xingyun Liu
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yong Li
- The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Sheng Huang
- Jiuzhitang Co., Ltd, Changsha, Hunan 410007, China
| | - Zhe Wang
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China.
| | - Guotao Tang
- Institute of Pharmacy and Pharmacology, Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medicial School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
3
|
Li Y, Zhang MZ, Zhang SJ, Sun X, Zhou C, Li J, Liu J, Feng J, Lu SY, Pei-Jun L, Wang JC. HIF-1α inhibitor YC-1 suppresses triple-negative breast cancer growth and angiogenesis by targeting PlGF/VEGFR1-induced macrophage polarization. Biomed Pharmacother 2023; 161:114423. [PMID: 36822023 DOI: 10.1016/j.biopha.2023.114423] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/23/2023] Open
Abstract
Triple negative breast cancer (TNBC) is an invasive and metastatic phenotype of breast cancer with limited treatment options. Published studies have demonstrated an inhibitory effect of HIF-α inhibition by its inhibitor YC-1 (lificiguat) on growth and angiogenesis of TNBC. However, the underlying mechanism remains poorly understood. In the current paper, our results show that HIF-1α inhibitor significantly inhibited TNBC growth by increasing cellular apoptosis and decreasing MVD, independent of a cell-autonomous mechanism in both endothelial and tumor cells. Genetic screening and in vivo experiments showed that a large number of M2-polarized TAMs accumulated in the hypoxic peri-necrotic region (PNR), where placental growth factor (PlGF) and its ligand, vascular endothelial growth factor receptor-1 (VEGFR-1) were upregulated. Furthermore, YC-1 skewed the polarization of TAMs away from M2 to M1 phenotype, therefore inhibiting TNBC angiogenesis and growth. This effect was further abrogated by VEGFR-1 neutralization and TAM depletion following clodronate liposome injection. These findings provide preclinical evidence for an indirect mechanism underlying YC-1-induced suppression of TNBC growth and angiogenesis, thereby offering a treatment option for TNBC.
Collapse
Affiliation(s)
- Yan Li
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Meng-Zhao Zhang
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Shu-Jing Zhang
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Xin Sun
- Department of Thoracic Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Can Zhou
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Juan Li
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, First Affiliated Hospital of Xi'an Jiaotong University, 710061, China
| | - Jie Liu
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, First Affiliated Hospital of Xi'an Jiaotong University, 710061, China
| | - Jun Feng
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Shao-Ying Lu
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China
| | - Liu Pei-Jun
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, First Affiliated Hospital of Xi'an Jiaotong University, 710061, China.
| | - Ji-Chang Wang
- Department of Vascular Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710061, China.
| |
Collapse
|
4
|
Yu KH, Hung HY. Synthetic strategy and structure-activity relationship (SAR) studies of 3-(5'-hydroxymethyl-2'-furyl)-1-benzyl indazole (YC-1, Lificiguat): a review. RSC Adv 2021; 12:251-264. [PMID: 35424505 PMCID: PMC8978903 DOI: 10.1039/d1ra08120a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/14/2021] [Indexed: 01/04/2023] Open
Abstract
Since 1994, YC-1 (Lificiguat, 3-(5′-hydroxymethyl-2′-furyl)-1-benzylindazole) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of hypoxia-inducible factor-1 (HIF-1) and NF-κB. Recently, Riociguat®, the first soluble guanylate cyclase (sGC) stimulator drug used to treat pulmonary hypertension and pulmonary arterial hypertension, was derived from the YC-1 structure. In this review, we aim to highlight the synthesis and structure–activity relationships in the development of YC-1 analogs and their possible indications. Since 1994, YC-1 (Lificiguat) has been synthesized, and many targets for special bioactivities have been explored, such as stimulation of platelet-soluble guanylate cyclase, indirect elevation of platelet cGMP levels, and inhibition of HIF-1 and NF-κB.![]()
Collapse
Affiliation(s)
- Ko-Hua Yu
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| | - Hsin-Yi Hung
- School of Pharmacy College of Medicine, National Cheng Kung University Tainan 701 Taiwan
| |
Collapse
|
5
|
Guan XY, Guan XL, Jiao ZY. Improving therapeutic resistance: beginning with targeting the tumor microenvironment. J Chemother 2021; 34:492-516. [PMID: 34873999 DOI: 10.1080/1120009x.2021.2011661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Cancer is a serious threat to human health and life. The tumor microenvironment (TME) not only plays a key role in the occurrence, development and metastasis of cancer, but also has a profound impact on treatment resistance. To improve and solve this problem, an increasing number of strategies targeting the TME have been proposed, and great progress has been made in recent years. This article reviews the characteristics and functions of the main matrix components of the TME and the mechanisms by which each component affects drug resistance. Furthermore, this article elaborates on targeting the TME as a strategy to treat acquired drug resistance, reduce tumor metastasis, recurrence, and improve efficacy.
Collapse
Affiliation(s)
- Xiao-Ying Guan
- Pathology Department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiao-Li Guan
- General Medicine Department, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zuo-Yi Jiao
- The First Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
6
|
Xia Z, Xiao J, Chen Q. Solving the Puzzle: What Is the Role of Progestogens in Neovascularization? Biomolecules 2021; 11:1686. [PMID: 34827682 PMCID: PMC8615949 DOI: 10.3390/biom11111686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
Ovarian sex steroids can modulate new vessel formation and development, and the clarification of the underlying mechanism will provide insight into neovascularization-related physiological changes and pathological conditions. Unlike estrogen, which mainly promotes neovascularization through activating classic post-receptor signaling pathways, progesterone (P4) regulates a variety of downstream factors with angiogenic or antiangiogenic effects, exerting various influences on neovascularization. Furthermore, diverse progestins, the synthetic progesterone receptor (PR) agonists structurally related to P4, have been used in numerous studies, which could contribute to unequal actions. As a result, there have been many conflicting observations in the past, making it difficult for researchers to define the exact role of progestogens (PR agonists including naturally occurring P4 and synthetic progestins). This review summarizes available evidence for progestogen-mediated neovascularization under physiological and pathological circumstances, and attempts to elaborate their functional characteristics and regulatory patterns from a comprehensive perspective.
Collapse
Affiliation(s)
| | | | - Qiong Chen
- Department of Geriatrics, Xiangya Hospital of Central South University, Changsha 410008, China; (Z.X.); (J.X.)
| |
Collapse
|
7
|
Xiao S, Li Q, Hu L, Yu Z, Yang J, Chang Q, Chen Z, Hu G. Soluble Guanylate Cyclase Stimulators and Activators: Where are We and Where to Go? Mini Rev Med Chem 2019; 19:1544-1557. [PMID: 31362687 DOI: 10.2174/1389557519666190730110600] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/05/2019] [Accepted: 04/20/2019] [Indexed: 02/04/2023]
Abstract
Soluble Guanylate Cyclase (sGC) is the intracellular receptor of Nitric Oxide (NO). The activation of sGC results in the conversion of Guanosine Triphosphate (GTP) to the secondary messenger cyclic Guanosine Monophosphate (cGMP). cGMP modulates a series of downstream cascades through activating a variety of effectors, such as Phosphodiesterase (PDE), Protein Kinase G (PKG) and Cyclic Nucleotide-Gated Ion Channels (CNG). NO-sGC-cGMP pathway plays significant roles in various physiological processes, including platelet aggregation, smooth muscle relaxation and neurotransmitter delivery. With the approval of an sGC stimulator Riociguat for the treatment of Pulmonary Arterial Hypertension (PAH), the enthusiasm in the discovery of sGC modulators continues for broad clinical applications. Notably, through activating the NO-sGC-cGMP pathway, sGC stimulator and activator potentiate for the treatment of various diseases, such as PAH, Heart Failure (HF), Diabetic Nephropathy (DN), Systemic Sclerosis (SS), fibrosis as well as other diseases including Sickle Cell Disease (SCD) and Central Nervous System (CNS) disease. Here, we review the preclinical and clinical studies of sGC stimulator and activator in recent years and prospect for the development of sGC modulators in the near future.
Collapse
Affiliation(s)
- Sijia Xiao
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Liqing Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Zutao Yu
- Department of Chemistry, Graduate School of Science Kyoto University Kitashirakawa- Oiwakecho, Sakyo-Ku, kyoto, Japan
| | - Jie Yang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Qi Chang
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Gaoyun Hu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| |
Collapse
|
8
|
DiGiacomo JW, Gilkes DM. Tumor Hypoxia As an Enhancer of Inflammation-Mediated Metastasis: Emerging Therapeutic Strategies. Target Oncol 2019; 13:157-173. [PMID: 29423593 DOI: 10.1007/s11523-018-0555-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Metastasis is the leading cause of cancer-related deaths. Recent research has implicated tumor inflammation as a promoter of metastasis. Myeloid, lymphoid, and mesenchymal cells in the tumor microenvironment promote inflammatory signaling amongst each other and together with cancer cells to modulate sustained inflammation, which may enhance cancer invasiveness. Tumor hypoxia, a state of reduced available oxygen present in the majority of solid tumors, acts as a prognostic factor for a worse outcome and is known to have a role in tumor inflammation through the regulation of inflammatory mediator signals in both cancer and neighboring cells in the microenvironment. Multiple methods to target tumor hypoxia have been developed and tested in clinical trials, and still more are emerging as the impacts of hypoxia become better understood. These strategies include mechanistic inhibition of the hypoxia inducible factor signaling pathway and hypoxia activated pro-drugs, leading to both anti-tumor and anti-inflammatory effects. This prompts a need for further research on the prevention of hypoxia-mediated inflammation in cancer. Hypoxia-targeting strategies seem to have the most potential for therapeutic benefit when combined with traditional chemotherapy agents. This paper will serve to summarize the role of the inflammatory response in metastasis, to discuss how hypoxia can enable or enhance inflammatory signaling, and to review established and emerging strategies to target the hypoxia-inflammation-metastasis axis.
Collapse
Affiliation(s)
- Josh W DiGiacomo
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.,Breast & Ovarian Cancer Program, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Daniele M Gilkes
- Department of Chemical & Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA. .,Breast & Ovarian Cancer Program, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
9
|
DiGiacomo JW, Gilkes DM. Therapeutic Strategies to Block the Hypoxic Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:141-157. [DOI: 10.1007/978-3-030-12734-3_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
10
|
Fujita H, Hirose K, Sato M, Fujioka I, Fujita T, Aoki M, Takai Y. Metformin attenuates hypoxia-induced resistance to cisplatin in the HepG2 cell line. Oncol Lett 2018; 17:2431-2440. [PMID: 30719114 DOI: 10.3892/ol.2018.9869] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/14/2018] [Indexed: 12/30/2022] Open
Abstract
Hepatoblastoma is the most commonly occurring liver tumor in children. Preoperative chemotherapy and surgery have improved treatment outcomes; however, further improvements are required in the treatment of advanced cases. Recently, the efficacy of transarterial chemoembolization (TACE) has garnered attention. TACE increases the local concentration of drugs by transcatheterically administering antitumor agents, and induces necrosis in the tumor by embolizing the feeding artery. However, studies have revealed that tumors exhibit resistance to anticancer drugs in hypoxic environments. Metformin is a drug used to treat type 2 diabetes; however, recent reports have indicated that it may also exhibit antitumor effects in various cancer cell lines. These effects are hypothesized to be mediated by the activation of adenosine monophosphate-activated protein kinase and reduction of mammalian target of rapamycin signaling, but these effects occur at high concentrations that are not suitable for use in a clinical setting. The potential efficacy of metformin at increased physiological concentrations has not been evaluated. The present study investigated the therapeutic effect of low concentrations of metformin in combination with cisplatin on liver cancer HepG2 cells in hypoxic conditions. HepG2 cells were treated with cisplatin alone, metformin alone, or a combination of these two drugs and cultured in normoxia or hypoxia. Treatment with either 5 µM cisplatin or 1 mM metformin alone did not significantly affect cell proliferation or apoptosis in hypoxic conditions. However, when 5 µM cisplatin was combined with 1 mM metformin, a significant inhibition of cell proliferation and induction of apoptosis was observed in hypoxic HepG2 cells. In conclusion, a low concentration of metformin attenuates hypoxia-induced resistance to cisplatin in HepG2 cells. Selective delivery of an effective dose of metformin to a hepatoblastoma tumor may be achievable and clinically useful with TACE.
Collapse
Affiliation(s)
- Hiromasa Fujita
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Katsumi Hirose
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan.,Department of Radiation Oncology, Southern Tohoku BNCT Research Center, Koriyama, Fukushima 963-8052, Japan
| | - Mariko Sato
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Ichitaro Fujioka
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Tamaki Fujita
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Masahiko Aoki
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Yoshihiro Takai
- Department of Radiology and Radiation Oncology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan.,Department of Radiation Oncology, Southern Tohoku BNCT Research Center, Koriyama, Fukushima 963-8052, Japan
| |
Collapse
|
11
|
Liang Y, Mafuvadze B, Besch-Williford C, Hyder SM. A combination of p53-activating APR-246 and phosphatidylserine-targeting antibody potently inhibits tumor development in hormone-dependent mutant p53-expressing breast cancer xenografts. BREAST CANCER-TARGETS AND THERAPY 2018; 10:53-67. [PMID: 29606888 PMCID: PMC5868596 DOI: 10.2147/bctt.s156285] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background Between 30 and 40% of human breast cancers express a defective tumor suppressor p53 gene. Wild-type p53 tumor suppressor protein promotes cell-cycle arrest and apoptosis and inhibits vascular endothelial growth factor-dependent angiogenesis, whereas mutant p53 protein (mtp53) lacks these functions, resulting in tumor cell survival and metastasis. Restoration of p53 function is therefore a promising drug-targeted strategy for combating mtp53-expressing breast cancer. Methods In this study, we sought to determine whether administration of APR-246, a small-molecule drug that restores p53 function, in combination with 2aG4, an antibody that targets phosphatidylserine residues on tumor blood vessels and disrupts tumor vasculature, effectively inhibits advanced hormone-dependent breast cancer tumor growth. Results APR-246 reduced cell viability in mtp53-expressing BT-474 and T47-D human breast cancer cells in vitro, and significantly induced apoptosis in a dose-dependent manner. However, APR-246 did not reduce cell viability in MCF-7 breast cancer cells, which express wild-type p53. We next examined APR-246's anti-tumor effects in vivo using BT-474 and T47-D tumor xenografts established in female nude mice. Tumor-bearing mice were treated with APR-246 and/or 2aG4 and tumor volume followed over time. Tumor growth was more effectively suppressed by combination treatment than by either agent alone, and combination therapy completely eradicated some tumors. Immunohistochemistry analysis of tumor tissue sections demonstrated that combination therapy more effectively induced apoptosis and reduced cell proliferation in tumor xenografts than either agent alone. Importantly, combination therapy dramatically reduced the density of blood vessels, which serve as the major route for tumor metastasis, in tumor xenografts compared with either agent alone. Conclusion Based on our findings, we contend that breast tumor growth might effectively be controlled by simultaneous targeting of mtp53 protein and tumor blood vessels in mtp53-expressing cancers.
Collapse
Affiliation(s)
- Yayun Liang
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| | - Benford Mafuvadze
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| | | | - Salman M Hyder
- Deparment of Biomedical Sciences and Dalton Cardiovascular Research Center, Columbia, MO, USA
| |
Collapse
|
12
|
Saberi-Karimian M, Katsiki N, Caraglia M, Boccellino M, Majeed M, Sahebkar A. Vascular endothelial growth factor: An important molecular target of curcumin. Crit Rev Food Sci Nutr 2017; 59:299-312. [DOI: 10.1080/10408398.2017.1366892] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maryam Saberi-Karimian
- Student Research Committee, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, University of Campania “L. Vanvitelli” 7, Via L. De Crecchio 7, Naples, Italy
| | - Mariarosaria Boccellino
- Department of Biochemistry, Biophysics and General Pathology, University of Campania “L. Vanvitelli” 7, Via L. De Crecchio 7, Naples, Italy
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Lee MR, Lin C, Lu CC, Kuo SC, Tsao JW, Juan YN, Chiu HY, Lee FY, Yang JS, Tsai FJ. YC-1 induces G 0/G 1 phase arrest and mitochondria-dependent apoptosis in cisplatin-resistant human oral cancer CAR cells. Biomedicine (Taipei) 2017; 7:12. [PMID: 28612710 PMCID: PMC5479426 DOI: 10.1051/bmdcn/2017070205] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/02/2017] [Indexed: 12/15/2022] Open
Abstract
Oral cancer is a serious and fatal disease. Cisplatin is the first line of chemotherapeutic agent for oral cancer therapy. However, the development of drug resistance and severe side effects cause tremendous problems clinically. In this study, we investigated the pharmacologic mechanisms of YC-1 on cisplatin-resistant human oral cancer cell line, CAR. Our results indicated that YC-1 induced a concentration-dependent and time-dependent decrease in viability of CAR cells analyzed by MTT assay. Real-time image analysis of CAR cells by IncuCyte™ Kinetic Live Cell Imaging System demonstrated that YC-1 inhibited cell proliferation and reduced cell confluence in a time-dependent manner. Results from flow cytometric analysis revealed that YC-1 promoted G0/G1 phase arrest and provoked apoptosis in CAR cells. The effects of cell cycle arrest by YC-1 were further supported by up-regulation of p21 and down-regulation of cyclin A, D, E and CDK2 protein levels. TUNEL staining showed that YC-1 caused DNA fragmentation, a late stage feature of apoptosis. In addition, YC-1 increased the activities of caspase-9 and caspase-3, disrupted the mitochondrial membrane potential (AYm) and stimulated ROS production in CAR cells. The protein levels of cytochrome c, Bax and Bak were elevated while Bcl-2 protein expression was attenuated in YC-1-treated CAR cells. In summary, YC-1 suppressed the viability of cisplatin-resistant CAR cells through inhibiting cell proliferation, arresting cell cycle at G0/G1 phase and triggering mitochondria-mediated apoptosis. Our results provide evidences to support the potentially therapeutic application of YC-1 on fighting against drug resistant oral cancer in the future.
Collapse
Affiliation(s)
- Miau-Rong Lee
- Department of Biochemistry, China Medical University, Taichung 404, Taiwan
| | - Chingju Lin
- Department of Physiology, China Medical University, Taichung 404, Taiwan
| | - Chi-Cheng Lu
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan - Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Sheng-Chu Kuo
- Chinese Medicinal Research and Development Center, China Medical University Hospital, China Medical University, Taichung 404, Taiwan - School of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Je-Wei Tsao
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Yu-Ning Juan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Hong-Yi Chiu
- Department of Pharmacy, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Fang-Yu Lee
- Yung-Shin Pharmaceutical Industry Co., Ltd., Tachia, Taichung 437, Taiwan
| | - Jai-Sing Yang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Fuu-Jen Tsai
- Genetics Center, Department of Medical Research, China Medical University Hospital, Taichung 404, Taiwan - School of Chinese Medicine, China Medical University, Taichung 404, Taiwan - Department of Medical Genetics, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
14
|
Alvarado A, Faustino-Rocha AI, Colaço B, Oliveira PA. Experimental mammary carcinogenesis - Rat models. Life Sci 2017; 173:116-134. [PMID: 28188729 DOI: 10.1016/j.lfs.2017.02.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 01/26/2017] [Accepted: 02/06/2017] [Indexed: 12/22/2022]
Abstract
Mammary cancer is one of the most common cancers, victimizing more than half a million of women worldwide every year. Despite all the studies in this field, the current therapeutic approaches are not effective and have several devastating effects for patients. In this way, the need to better understand the mammary cancer biopathology and find effective therapies led to the development of several rodent models over years. With this review, the authors intended to provide the readers with an overview of the rat models used to study mammary carcinogenesis, with a special emphasis on chemically-induced models.
Collapse
Affiliation(s)
- Antonieta Alvarado
- Área de Patología, Decanato de Ciencias Veterinarias, Universidad Centroccidental "Lisandro Alvarado", UCLA, Lara, Venezuela; Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal
| | - Ana I Faustino-Rocha
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Bruno Colaço
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Zootechnics, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal
| | - Paula A Oliveira
- Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences, UTAD, Vila Real, Portugal.
| |
Collapse
|
15
|
Wang X, Liu C, Wu L, Zhu S. Potent ameliorating effect of Hypoxia-inducible factor 1α (HIF-1α) antagonist YC-1 on combined allergic rhinitis and asthma syndrome (CARAS) in Rats. Eur J Pharmacol 2016; 788:343-350. [PMID: 27498367 DOI: 10.1016/j.ejphar.2016.07.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/21/2016] [Accepted: 07/22/2016] [Indexed: 12/25/2022]
Abstract
Recent studies have implicated that Hypoxia-inducible factor 1α (HIF-1α) plays an integral role in the pathogenesis of allergic rhinitis and asthma. In the present study, we showed that HIF-1α antagonist YC-1, 3-(5-hydroxymethyl-2-furyl)-1-benzylindazole, elicited a potent allergy-ameliorating effect in a rat model of ovalbumin (OVA)-sensitized combined allergic rhinitis and asthma syndrome (CARAS). We revealed that YC-1 administration markedly impaired the total number and percentage of eosinophil in bronchoalveolar lavage fluid (BAL Fluid) of the rats, suggesting that YC-1 might attenuate lung and nasal mucosal inflammation in OVA-sensitized rats. Moreover, histological examination found that OVA-induced pathological alterations were evidently attenuated following YC-1 administration. In addition, immunohistochemistrial analysis indicated that YC-1 treatment decreased the expression of HIF-1α in rat lungs and nasal mucosa. Notably, Nuclear factor kappa B (NF-κB) p65 and Peroxisome proliferator-activated receptor α (PPARα), two important regulators of inflammatory responses, were also significantly down-regulated following YC-1 administration. Real-time PCR analysis confirmed that YC-1 impaired the expression of HIF-1α, NF-κB and PPARα in CARAS model. These findings together indicated that YC-1 exerted remarkable anti-allergic effects through the modulation of inflammatory pathways, implying that YC-1 may potentially serve as a novel anti-CARAS medicine in clinical patients.
Collapse
Affiliation(s)
- Xu Wang
- Laboratory Animals Center, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Chun Liu
- Laboratory Animals Center, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Liucheng Wu
- Laboratory Animals Center, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | - Shunxing Zhu
- Laboratory Animals Center, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China.
| |
Collapse
|
16
|
COOK MATTHEWT, MAFUVADZE BENFORD, BESCH-WILLIFORD CYNTHIA, ELLERSIECK MARKR, GOYETTE SANDY, HYDER SALMANM. Luteolin suppresses development of medroxyprogesterone acetate-accelerated 7,12-dimethylbenz(a)anthracene-induced mammary tumors in Sprague-Dawley rats. Oncol Rep 2015; 35:825-32. [DOI: 10.3892/or.2015.4431] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 10/23/2015] [Indexed: 11/06/2022] Open
|
17
|
Botelho MC, Soares R, Alves H. Progesterone in Breast Cancer Angiogenesis. SM JOURNAL OF REPRODUCTIVE HEALTH & INFERTILITY 2015; 1:1001. [PMID: 26640826 PMCID: PMC4667742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The involvement of steroid hormones in breast carcinogenesis is well established. Recent evidence suggests that angiogenesis can be regulated by hormones. Both oestrogen and progesterone have been implicated in the angiogenic process of hormone-dependent cancers, such as breast cancer. Vascular Endothelial Growth Factor (VEGF) is a growth factor involved in angiogenesis in breast cancer that is up-regulated by estrogens. In our study we evaluated the role of progesterone in the expression of this angiogenic growth factor commonly up-regulated in breast cancer. Our findings indicate that progesterone activates an angiogenic pathway involving VEGF stimulation. The elucidation of specific angiogenic pathways promoted by progesterone can raise new therapeutic targets at least in a subset of breast cancers responsive to progesterone.
Collapse
Affiliation(s)
- Monica C Botelho
- Unit of Health Promotion and Chronic Diseases, National Institute of Health (INSA) Dr. Ricardo Jorge, Porto, Portugal
| | - Raquel Soares
- IPATIMUP, Institute of Pathology and Molecular Immunology of the University of Porto, Portugal
| | - Helena Alves
- I3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| |
Collapse
|
18
|
Chen J, Wang J, Schwab LP, Park KT, Seagroves TN, Jennings LK, Miller DD, Li W. Benzimidazole analogs as potent hypoxia inducible factor inhibitors: synthesis, biological evaluation, and profiling drug-like properties. Anticancer Res 2014; 34:3891-3904. [PMID: 25075010 PMCID: PMC5346463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
AIM To develop potent HIF-1α inhibitors for potential treatment of cancer. MATERIALS AND METHODS Chemical synthesis, HIF-luciferase assay, cytotoxic assay, platelet aggregation assay, western blot analysis, quantitative real-time PCR, aqueous solubility, protein binding, metabolic stability, and metabolic pathways. RESULTS Thirteen novel benzimidazole analogs were synthesized. Compounds 3a and 3k showed the highest anti-HIF-1α activity. They are significantly more effective than YC-1 in the suppression of HIF-1α protein expression based on western blot assay. They show comparable potency in inhibition of cancer cell migration. They are less potent in the inhibition of platelet aggregation. 3k had the most favorable drug-like properties, including long half-life in human liver microsomes, medium protein binding level and reasonable aqueous solubility. CONCLUSION The potent anti-HIF-1α activity and favorable drug-like properties of compound 3k suggest that it may hold great potential as an adjuvant therapy for cancer treatment through repression of HIF-1α protein expression.
Collapse
Affiliation(s)
- Jianjun Chen
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, U.S.A. Department of Pharmaceutical Sciences, School of Pharmacy, South College, Knoxville, TN, U.S.A
| | - Jin Wang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Luciana P Schwab
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Kyung-Tae Park
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Tiffany N Seagroves
- Center for Cancer Research, Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Lisa K Jennings
- Vascular Biology Center, Department of Internal Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Duane D Miller
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, U.S.A.
| |
Collapse
|