1
|
Jain SM, Nagainallur Ravichandran S, Murali Kumar M, Banerjee A, Sun-Zhang A, Zhang H, Pathak R, Sun XF, Pathak S. Understanding the molecular mechanism responsible for developing therapeutic radiation-induced radioresistance of rectal cancer and improving the clinical outcomes of radiotherapy - A review. Cancer Biol Ther 2024; 25:2317999. [PMID: 38445632 PMCID: PMC10936619 DOI: 10.1080/15384047.2024.2317999] [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: 08/16/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
Rectal cancer accounts for the second highest cancer-related mortality, which is predominant in Western civilizations. The treatment for rectal cancers includes surgery, radiotherapy, chemotherapy, and immunotherapy. Radiotherapy, specifically external beam radiation therapy, is the most common way to treat rectal cancer because radiation not only limits cancer progression but also significantly reduces the risk of local recurrence. However, therapeutic radiation-induced radioresistance to rectal cancer cells and toxicity to normal tissues are major drawbacks. Therefore, understanding the mechanistic basis of developing radioresistance during and after radiation therapy would provide crucial insight to improve clinical outcomes of radiation therapy for rectal cancer patients. Studies by various groups have shown that radiotherapy-mediated changes in the tumor microenvironment play a crucial role in developing radioresistance. Therapeutic radiation-induced hypoxia and functional alterations in the stromal cells, specifically tumor-associated macrophage (TAM) and cancer-associated fibroblasts (CAF), play a crucial role in developing radioresistance. In addition, signaling pathways, such as - the PI3K/AKT pathway, Wnt/β-catenin signaling, and the hippo pathway, modulate the radiation responsiveness of cancer cells. Different radiosensitizers, such as small molecules, microRNA, nanomaterials, and natural and chemical sensitizers, are being used to increase the effectiveness of radiotherapy. This review highlights the mechanism responsible for developing radioresistance of rectal cancer following radiotherapy and potential strategies to enhance the effectiveness of radiotherapy for better management of rectal cancer.
Collapse
Affiliation(s)
- Samatha M Jain
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Shruthi Nagainallur Ravichandran
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Makalakshmi Murali Kumar
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| | - Alexander Sun-Zhang
- Department of Oncology-Pathology, BioClinicum, Karolinska Institutet, Stockholm, Sweden
| | - Hong Zhang
- School of Medicine, Department of Medical Sciences, Orebro University, Örebro, Sweden
| | - Rupak Pathak
- Division of Radiation Health, Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam, Chennai, India
| |
Collapse
|
2
|
Ali Beg MM, Saxena A, Singh VK, Akhter J, Habib H, Raisuddin S. Modulatory role of BV6 and chloroquine on the regulation of apoptosis and autophagy in non-small cell lung cancer cells. J Cancer Res Ther 2023; 19:S0. [PMID: 37147964 DOI: 10.4103/jcrt.jcrt_816_21] [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: 05/07/2023]
Abstract
Aims Non-small cell lung cancer (NSCLC) is one of the aggressive tumors mostly diagnosed in the advanced stage. Therapeutic failure and drug resistance pose a major problem in NSCLC treatment primarily due to alterations in autophagy and loss of apoptosis. Therefore, the present study aimed to investigate the importance of the second mitochondria-derived activator of caspase mimetic BV6 and autophagy inhibitor chloroquine (CQ) on the regulation of apoptosis and autophagy, respectively. Subjects and Methods Study was conducted on NCI-H23 and NCI-H522 cell lines to evaluate the effect of BV6 and CQ on the transcription and translation level of LC3-II, caspase-3, and caspase-9 genes by quantitative real-time-polymerase chain reaction and western blotting techniques. Results In NCI-H23 cell line, BV6 and CQ treatments showed increased mRNA and protein expression of caspase-3, and caspase-9 compared to its untreated counterpart. BV6 and CQ treatments also caused downregulation of LC3-II protein expression compared to its counterpart. In NCI-H522 cell line, BV6 treatment showed a significantly increased expression of caspase-3 and caspase-9 mRNA and protein expression levels whereas BV6 treatment downregulated the expression level of LC3-II protein. A similar pattern was also observed in CQ treatment when compared with the respective controls. Both BV6 and CQ modulated in vitro expression of caspases and LC3-II which have critical regulatory roles in apoptosis and autophagy, respectively. Conclusions Our findings suggest that BV6 and CQ could be promising candidates in NSCLC treatment and there is a need to explore them in vivo and in clinical applications.
Collapse
Affiliation(s)
- Mirza Masroor Ali Beg
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India; Biochemistry, Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyzstan
| | - Alpana Saxena
- Biochemistry, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard (Hamdard University), New Delhi, India
| | | | - Juheb Akhter
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Haroon Habib
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Sheikh Raisuddin
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
3
|
Sun H, Liu F, Zhai H, Wu J, Nie S, Cai H, Wen K, Feng L, Liu Q, Ji K, Wang Y. Self-synthesized second mitochondria-derived activator of caspase (SMAC) mimetic TP-WY-1345 enhances the radiosensitivity of NSCLC cells H1299 by targeting anti-apoptotic protein cIAP1. RADIATION MEDICINE AND PROTECTION 2023. [DOI: 10.1016/j.radmp.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
4
|
Short Linear Motifs in Colorectal Cancer Interactome and Tumorigenesis. Cells 2022; 11:cells11233739. [PMID: 36496998 PMCID: PMC9737320 DOI: 10.3390/cells11233739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Colorectal tumorigenesis is driven by alterations in genes and proteins responsible for cancer initiation, progression, and invasion. This multistage process is based on a dense network of protein-protein interactions (PPIs) that become dysregulated as a result of changes in various cell signaling effectors. PPIs in signaling and regulatory networks are known to be mediated by short linear motifs (SLiMs), which are conserved contiguous regions of 3-10 amino acids within interacting protein domains. SLiMs are the minimum sequences required for modulating cellular PPI networks. Thus, several in silico approaches have been developed to predict and analyze SLiM-mediated PPIs. In this review, we focus on emerging evidence supporting a crucial role for SLiMs in driver pathways that are disrupted in colorectal cancer (CRC) tumorigenesis and related PPI network alterations. As a result, SLiMs, along with short peptides, are attracting the interest of researchers to devise small molecules amenable to be used as novel anti-CRC targeted therapies. Overall, the characterization of SLiMs mediating crucial PPIs in CRC may foster the development of more specific combined pharmacological approaches.
Collapse
|
5
|
Townsend PA, Kozhevnikova MV, Cexus ONF, Zamyatnin AA, Soond SM. BH3-mimetics: recent developments in cancer therapy. J Exp Clin Cancer Res 2021; 40:355. [PMID: 34753495 PMCID: PMC8576916 DOI: 10.1186/s13046-021-02157-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/26/2021] [Indexed: 01/11/2023] Open
Abstract
The hopeful outcomes from 30 years of research in BH3-mimetics have indeed served a number of solid paradigms for targeting intermediates from the apoptosis pathway in a variety of diseased states. Not only have such rational approaches in drug design yielded several key therapeutics, such outputs have also offered insights into the integrated mechanistic aspects of basic and clinical research at the genetics level for the future. In no other area of medical research have the effects of such work been felt, than in cancer research, through targeting the BAX-Bcl-2 protein-protein interactions. With these promising outputs in mind, several mimetics, and their potential therapeutic applications, have also been developed for several other pathological conditions, such as cardiovascular disease and tissue fibrosis, thus highlighting the universal importance of the intrinsic arm of the apoptosis pathway and its input to general tissue homeostasis. Considering such recent developments, and in a field that has generated so much scientific interest, we take stock of how the broadening area of BH3-mimetics has developed and diversified, with a focus on their uses in single and combined cancer treatment regimens and recently explored therapeutic delivery methods that may aid the development of future therapeutics of this nature.
Collapse
Affiliation(s)
- Paul A Townsend
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
- University of Manchester, Manchester, UK.
| | - Maria V Kozhevnikova
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
| | | | - Andrey A Zamyatnin
- University of Surrey, Guildford, UK
- Sechenov First Moscow State Medical University, Moscow, Russian Federation
- Lomonosov Moscow State University, Moscow, Russian Federation
- Sirius University of Science and Technology, Sochi, Russian Federation
| | - Surinder M Soond
- University of Surrey, Guildford, UK.
- Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| |
Collapse
|
6
|
Zhao XY, Wang XY, Wei QY, Xu YM, Lau ATY. Potency and Selectivity of SMAC/DIABLO Mimetics in Solid Tumor Therapy. Cells 2020; 9:cells9041012. [PMID: 32325691 PMCID: PMC7226512 DOI: 10.3390/cells9041012] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 02/05/2023] Open
Abstract
Aiming to promote cancer cell apoptosis is a mainstream strategy of cancer therapy. The second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis protein (IAP)-binding protein with low pI (DIABLO) protein is an essential and endogenous antagonist of inhibitor of apoptosis proteins (IAPs). SMAC mimetics (SMs) are a series of synthetically chemical compounds. Via database analysis and literature searching, we summarize the potential mechanisms of endogenous SMAC inefficiency, degradation, mutation, releasing blockage, and depression. We review the development of SMs, as well as preclinical and clinical outcomes of SMs in solid tumor treatment, and we analyze their strengths, weaknesses, opportunities, and threats from our point of view. We also highlight several questions in need of further investigation.
Collapse
Affiliation(s)
| | | | | | - Yan-Ming Xu
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| | - Andy T. Y. Lau
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| |
Collapse
|
7
|
Esmaeili Govarchin Ghaleh H, Zarei L, Mansori Motlagh B, Jabbari N. Using CuO nanoparticles and hyperthermia in radiotherapy of MCF-7 cell line: synergistic effect in cancer therapy. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1396-1403. [PMID: 30964344 DOI: 10.1080/21691401.2019.1600529] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The aim of this paper was examining the combined impacts of CuO nanoparticles (CuO NPs), hyperthermia (H), and irradiation (R) on an increment of MCF-7 cells. The MTT assay was employed to assess the antiproliferative effects of CuO NPs (25, 50, and 100 μg/ml), hyperthermia (41 °C for 1 h), and irradiation (200 cGy). Moreover, the perniciousness was estimated through the survival capability of cells, and apoptosis, ROS production, and levels of caspase-3, -8 and -9 proteins were determined. A significant (p < .01) decrease in proliferation index (0.124 ± 0.021), a significant (p < .01) increase in apoptosis (42% ± 1.54) of MCF7 cells, a significant (p < .03) increase in ROS formation (32.16 ± 1.9) and a significant (p < .01) increase in LDH release (33.28 ± 1.56) were recorded in the adjacency of MCF-7 cells by a combination of CuO NPs (100 µg/ml) and R + H compared to control and other treatments. The activities of caspase-3 (0.33 ± 0.014) and caspase-9 (0.389 ± 0.019) also increased significantly (p < .05). However, caspase-8 showed no significant changes in its activity (p = .065). Based on these observations, a combination of CuO NPs, hyperthermia, and irradiation could suppress the growth of MCF-7 cells and evoke cell apoptosis via mitochondrial membrane potential.
Collapse
Affiliation(s)
| | - Leila Zarei
- b Solid Tumor Research Center , Urmia University of Medical Sciences , Urmia , Iran
| | | | - Nasrollah Jabbari
- b Solid Tumor Research Center , Urmia University of Medical Sciences , Urmia , Iran
| |
Collapse
|
8
|
Yang L, Kumar B, Shen C, Zhao S, Blakaj D, Li T, Romito M, Teknos TN, Williams TM. LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus-negative Head and Neck Squamous Cell Carcinoma. Mol Cancer Ther 2019; 18:1025-1035. [PMID: 31015310 DOI: 10.1158/1535-7163.mct-18-1157] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/23/2019] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Targeting inhibitor of apoptosis proteins (IAP) with second mitochondria-derived activator of caspase (SMAC) mimetics may promote cancer cell death. We tested whether cIAP1 predicts poor prognosis in head and neck squamous cell carcinoma (HNSCC) and whether a novel Smac-mimetic, LCL161, could radiosensitize human papillomavirus-positive (HPV+) and -negative (HPV-) HNSCC. The association of BIRC2 (encoding cIAP1) mRNA level with HPV status in HNSCC was analyzed using The Cancer Genome Atlas (TCGA) database. cIAP1 was assessed by IHC on an HNSCC tissue microarray (TMA, n = 84) followed by correlation analysis with HPV status and patient outcomes. Human cell culture and animal models of HNSCC were used to analyze the outcome and molecular characteristics following radiotherapy in combination with LCL161. cIAP1 expression is increased in HPV- compared with HPV+HNSCC tumors in the TCGA database. In our TMA, cIAP1 was overexpressed in HNSCC compared with normal tissues (P = 0.0003) and associated with a poor overall survival (P = 0.0402). cIAP1 levels were higher in HPV- than that in HPV+HNSCC tumors (P = 0.004) and patients with cIAP1+/HPV- HNSCC had the worst survival. LCL161 effectively radiosensitized HPV- HNSCC cells, which was accompanied with enhanced apoptosis, but not HPV+ HNSCC cells. Importantly, LCL161 in combination with radiotherapy led to dramatic tumor regression of HPV- HNSCC tumor xenografts, accompanied by cIAP1 degradation and apoptosis activation. These results reveal that cIAP1 is a prognostic and a potential therapeutic biomarker for HNSCC, and targeting cIAP1 with LCL161 preferentially radiosensitizes HPV- HNSCC, providing justification for clinical testing of LCL161 in combination with radiation for patients with HPV- HNSCC.
Collapse
Affiliation(s)
- Linlin Yang
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Bhavna Kumar
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Changxian Shen
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Songzhu Zhao
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Dukagjin Blakaj
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Tianyun Li
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Mitchell Romito
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Theodoros N Teknos
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio
| | - Terence M Williams
- The Ohio State University Medical Center, Arthur G. James Comprehensive Cancer Center and Richard J. Solove Research Institute, Columbus, Ohio.
| |
Collapse
|
9
|
Hosseini F, Sam MR, Jabbari N, Mozdarani H. Modulating Survivin as a Radioresistant Factor, Caspase-3, and Apoptosis by Omega-3 Docosahexaenoic Acid Sensitizes Mutant-p53 Colorectal Cancer Cells to γ-Irradiation. Cancer Biother Radiopharm 2018; 33:387-395. [DOI: 10.1089/cbr.2018.2445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Affiliation(s)
- Farideh Hosseini
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Sam
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran
| | - Nasrollah Jabbari
- Department of Medical Physics and Imaging, Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Hossein Mozdarani
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
10
|
Liu B, Du L, Xu C, Wang Y, Wang Q, Song Z, Sun X, Wang J, Liu Q. [Radiosensitization Induced by ANTP-SmacN7 Fusion Peptide in H460 Cell Line]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2016; 19:241-6. [PMID: 27215450 PMCID: PMC5973049 DOI: 10.3779/j.issn.1009-3419.2016.05.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
背景与目的 肿瘤的辐射耐受制约了放疗疗效,第二个线粒体衍生的半胱氨酸蛋白酶激活剂(Second mitochondria-derived activator of caspase, Smac)蛋白类似物可明显提高辐射诱导的肿瘤细胞凋亡,有望成为新型肿瘤辐射增敏药物。本研究旨在探讨新型Smac蛋白类似物ANTP-SmacN7融合肽对肺癌细胞系H460的辐射增敏作用。 方法 合成ANTP-SmacN7融合肽,连接荧光素FITC以观察融合肽能否进入细胞。对数生长期H460细胞分为空白对照组、单纯照射组、ANTP-SmacN7组和照射联合ANTP-SmacN7组,单纯照射组给予0 Gy、2 Gy、4 Gy、6 Gy照射,照射联合ANTP-SmacN7组中ANTP-SmacN7的浓度为20 μmol/L,WST-1测定H460细胞的增殖。流式细胞仪测定细胞处理后24 h和48 h的细胞凋亡率。Western blot实验检测caspase3和cleaved caspase3的表达水平。 结果 ANTP-SmacN7融合能够顺利进入细胞,且能够增强H460细胞的辐射敏感性(F=25.1,P < 0.01,增敏比为1.86),照射联合ANTP-SmacN7可明显降低H460细胞的克隆形成率(χ2=45.2, P < 0.01; χ2=40.3, P < 0.01),提高cleaved caspase3的表达量,促进caspase3的活化,增加辐射诱导的细胞凋亡率。 结论 ANTP-SmacN7融合肽可明显提高H460细胞的辐射敏感性,作为一种新的Smac蛋白类似物有望用于肿瘤的辐射增敏治疗。
Collapse
Affiliation(s)
- Baona Liu
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Liqing Du
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Chang Xu
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Yan Wang
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Qin Wang
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Zhiyi Song
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Xiaohui Sun
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Jinhan Wang
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Qiang Liu
- Institute of Radiation Medicine of Chinese Academy of Medical Science and Peking Union Medical College, Tianjin Key Lab of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| |
Collapse
|
11
|
Goldar S, Khaniani MS, Derakhshan SM, Baradaran B. Molecular mechanisms of apoptosis and roles in cancer development and treatment. Asian Pac J Cancer Prev 2016; 16:2129-44. [PMID: 25824729 DOI: 10.7314/apjcp.2015.16.6.2129] [Citation(s) in RCA: 381] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Programmed cell death (PCD) or apoptosis is a mechanism which is crucial for all multicellular organisms to control cell proliferation and maintain tissue homeostasis as well as eliminate harmful or unnecessary cells from an organism. Defects in the physiological mechanisms of apoptosis may contribute to different human diseases like cancer. Identification of the mechanisms of apoptosis and its effector proteins as well as the genes responsible for apoptosis has provided a new opportunity to discover and develop novel agents that can increase the sensitivity of cancer cells to undergo apoptosis or reset their apoptotic threshold. These novel targeted therapies include those targeting anti-apoptotic Bcl-2 family members, p53, the extrinsic pathway, FLICE-inhibitory protein (c-FLIP), inhibitor of apoptosis (IAP) proteins, and the caspases. In recent years a number of these novel agents have been assessed in preclinical and clinical trials. In this review, we introduce some of the key regulatory molecules that control the apoptotic pathways, extrinsic and intrinsic death receptors, discuss how defects in apoptotic pathways contribute to cancer, and list several agents being developed to target apoptosis.
Collapse
Affiliation(s)
- Samira Goldar
- Department of Biochemistry and Clinical Labratorary, Division of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran E-mail :
| | | | | | | |
Collapse
|
12
|
Núñez de Villavicencio-Díaz T, Ramos Gómez Y, Oliva Argüelles B, Fernández Masso JR, Rodríguez-Ulloa A, Cruz García Y, Guirola-Cruz O, Perez-Riverol Y, Javier González L, Tiscornia I, Victoria S, Bollati-Fogolín M, Besada Pérez V, Guerra Vallespi M. Comparative proteomics analysis of the antitumor effect of CIGB-552 peptide in HT-29 colon adenocarcinoma cells. J Proteomics 2015; 126:163-71. [DOI: 10.1016/j.jprot.2015.05.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/06/2015] [Accepted: 05/19/2015] [Indexed: 10/25/2022]
|
13
|
Siddiqui MS, François M, Fenech MF, Leifert WR. Persistent γH2AX: A promising molecular marker of DNA damage and aging. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 766:1-19. [PMID: 26596544 DOI: 10.1016/j.mrrev.2015.07.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 07/13/2015] [Accepted: 07/14/2015] [Indexed: 12/12/2022]
Abstract
One of the earliest cellular responses to DNA double strand breaks (DSBs) is the phosphorylation of the core histone protein H2AX (termed γH2AX). Persistent γH2AX is the level of γH2AX above baseline, measured at a given time-point beyond which DNA DSBs are normally expected to be repaired (usually persist for days to months). This review summarizes the concept of persistent γH2AX in the context of exogenous source induced DNA DSBs (e.g. ionizing radiation (IR), chemotherapeutic drugs, genotoxic agents), and endogenous γH2AX levels in normal aging and accelerated aging disorders. Summary of the current literature demonstrates the following (i) γH2AX persistence is a common phenomenon that occurs in humans and animals; (ii) nuclei retain persistent γH2AX foci for up to several months after IR exposure, allowing for retrospective biodosimetry; (iii) the combination of various radiosensitizing drugs with ionizing radiation exposure leads to persistent γH2AX response, thus enabling the potential for monitoring cancer patients' response to chemotherapy and radiotherapy as well as tailoring cancer treatments; (iv) persistent γH2AX accumulates in telomeric DNA and in cells undergoing cellular senescence; and (v) increased endogenous γH2AX levels may be associated with diseases of accelerated aging. In summary, measurement of persistent γH2AX could potentially be used as a marker of radiation biodosimetry, evaluating sensitivity to therapeutic genotoxins and radiotherapy, and exploring the association of unrepaired DNA DSBs on telomeres with diseases of accelerated aging.
Collapse
Affiliation(s)
- Mohammad Sabbir Siddiqui
- CSIRO Food and Nutrition Flagship, Genome Health and Healthy Aging, Adelaide, South Australia 5000, Australia; University of Adelaide, School of Agriculture, Food & Wine, Urrbrae, South Australia 5064, Australia
| | - Maxime François
- CSIRO Food and Nutrition Flagship, Genome Health and Healthy Aging, Adelaide, South Australia 5000, Australia
| | - Michael F Fenech
- CSIRO Food and Nutrition Flagship, Genome Health and Healthy Aging, Adelaide, South Australia 5000, Australia
| | - Wayne R Leifert
- CSIRO Food and Nutrition Flagship, Genome Health and Healthy Aging, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
14
|
Matzinger O, Viertl D, Tsoutsou P, Kadi L, Rigotti S, Zanna C, Wiedemann N, Vozenin MC, Vuagniaux G, Bourhis J. The radiosensitizing activity of the SMAC-mimetic, Debio 1143, is TNFα-mediated in head and neck squamous cell carcinoma. Radiother Oncol 2015; 116:495-503. [PMID: 26096848 DOI: 10.1016/j.radonc.2015.05.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/26/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND PURPOSE Second mitochondria-derived activator of caspase (SMAC)-mimetics are a new class of targeted drugs that specifically induce apoptotic cancer cell death and block pro-survival signaling by antagonizing selected members of the inhibitor of apoptosis protein (IAP) family. MATERIAL AND METHODS The present study was designed to investigate the radiosensitizing effect and optimal sequence of administration of the novel SMAC-mimetic Debio 1143 in vitro and in vivo. Apoptosis, alteration of DNA damage repair (DDR), and tumor necrosis factor-alpha (TNF-α) signaling were examined. RESULTS In vitro, Debio 1143 displayed anti-proliferative activity and enhanced intrinsic radiation sensitivity in 5/6 head and neck squamous cell carcinoma (HNSCC) cell lines in a synergistic manner. In vivo, Debio 1143 dose-dependently radio-sensitized FaDu and SQ20B xenografts, resulting in complete tumor regression in 8/10 FaDu-xenografted mice at the high dose level. At the molecular level, Debio 1143 combined with radiotherapy (RT) induced enhancement of caspase-3 activity, increase in Annexin V-positive cells and karyopyknosis, and increase in TNF-α mRNA levels. Finally, in a neutralization experiment using a TNF-α-blocking antibody and a caspase inhibitor, it was shown that the radiosensitizing effect of Debio 1143 is mediated by caspases and TNF-α. CONCLUSIONS These results demonstrate that the novel SMAC-mimetic Debio 1143 is a radiosensitizing agent that is worthy of further investigation in clinical trials in combination with radiotherapy.
Collapse
Affiliation(s)
- Oscar Matzinger
- Laboratory of Radiation Oncology Department, University Hospital of Lausanne (CHUV), Switzerland; Radiation Oncology Department, Hôpital Riviera-Chablais, Vevey, Switzerland
| | - David Viertl
- Laboratory of Radiation Oncology Department, University Hospital of Lausanne (CHUV), Switzerland
| | - Pelagia Tsoutsou
- Laboratory of Radiation Oncology Department, University Hospital of Lausanne (CHUV), Switzerland; Radiation Oncology Department Hôpital du Valais, Sion, Switzerland
| | - Linda Kadi
- Debiopharm International SA, Lausanne, Switzerland
| | | | | | | | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology Department, University Hospital of Lausanne (CHUV), Switzerland.
| | | | - Jean Bourhis
- Laboratory of Radiation Oncology Department, University Hospital of Lausanne (CHUV), Switzerland
| |
Collapse
|
15
|
Flanagan L, Kehoe J, Fay J, Bacon O, Lindner AU, Kay EW, Deasy J, McNamara DA, Prehn JHM. High levels of X-linked Inhibitor-of-Apoptosis Protein (XIAP) are indicative of radio chemotherapy resistance in rectal cancer. Radiat Oncol 2015; 10:131. [PMID: 26071313 PMCID: PMC4480907 DOI: 10.1186/s13014-015-0437-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 06/05/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The mainstay of treatment in rectal cancer is neoadjuvant radio chemotherapy prior to surgery, in an attempt to downstage the tumour, allowing for more complete removal during surgery. In 40 % of cases however, this neoadjuvant radio chemotherapy fails to achieve tumour regression, partly due insufficient apoptosis signaling. X-linked Inhibitor of Apoptosis Protein (XIAP) is an anti-apoptotic protein that has been reported to contribute to disease progression and chemotherapy resistance. METHODS We obtained rectal biopsy normal and matched tumour tissue from 29 rectal cancer patients with varying degrees of tumour regression, and using Western blot, examined anti-apoptotic XIAP and pro-apoptotic Smac protein levels in these tissues, with the aim to examine whether disturbed XIAP/Smac levels may be an indicator of neoadjuvant radio chemotherapy resistance. Expression of inhibitor of apoptosis proteins cIAP-1 and cIAP-2 was also examined. RESULTS We found that levels of XIAP increased in accordance with the degree of radio chemotherapy resistance of the tissue. Levels of this protein were also significantly higher in tumour tissue, compared to matched normal tissue in highly resistant tissue. In contrast, Smac protein levels did not increase with radio chemotherapy resistance, and the protein was similarly expressed in normal and tumour tissue, indicating a shift in the balance of these proteins. Post treatment surgical resection tissue was available for 8 patients. When we compared matched tissue pre- and post- radio chemotherapy we found that XIAP levels increased significantly during treatment in both normal and tumour tissue, while Smac levels did not change. cIAP-1 and cIAP-2 levels were not differentially expressed in varying degrees of radio chemotherapy resistance, and neoadjuvant therapy did not alter expression of these proteins. CONCLUSION These data indicate that disturbance of the XIAP/Smac balance may be a driver of radio chemotherapy resistance, and hence high levels of XIAP may be a useful indicator of neoadjuvant radio chemotherapy resistance in rectal cancer. Moreover, as XIAP levels increase with radio chemotherapy it is possible that a subset of more resistant tumour cells survive this treatment and may be resistant to further adjuvant treatment. Patients with resistant tumours highly expressing XIAP may benefit from alternative treatment strategies, such as Smac mimetics post neoadjuvant radio chemotherapy.
Collapse
Affiliation(s)
- L Flanagan
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. .,Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - J Kehoe
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. .,Departments of Pathology, Beaumont Hospital, Dublin 9, Ireland. .,Departments of Surgery, Beaumont Hospital, Dublin 9, Ireland.
| | - J Fay
- Departments of Pathology, Beaumont Hospital, Dublin 9, Ireland.
| | - O Bacon
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - A U Lindner
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. .,Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | - E W Kay
- Departments of Pathology, Beaumont Hospital, Dublin 9, Ireland.
| | - J Deasy
- Departments of Surgery, Beaumont Hospital, Dublin 9, Ireland.
| | - D A McNamara
- Departments of Surgery, Beaumont Hospital, Dublin 9, Ireland.
| | - J H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland. .,Centre for Systems Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
16
|
Illum H, Wang DH, Dowell JE, Hittson WJ, Torrisi JR, Meyer J, Huerta S. Phase I dose escalation trial of nitroglycerin in addition to 5-fluorouracil and radiation therapy for neoadjuvant treatment of operable rectal cancer. Surgery 2015; 158:460-5. [PMID: 25964028 DOI: 10.1016/j.surg.2015.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 01/27/2015] [Accepted: 04/10/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND Nitric oxide donors decreased cell survival in vitro and tumor load in vivo in models of rectal cancer subjected to ionizing radiation. Nitroglycerin (NTG) transdermal patches, added to chemotherapy, have been shown to improve outcomes in lung cancer patients. METHODS This open-label, nonrandomized, multicohort, dose escalation, phase I trial had a primary endpoint to evaluate the safety, tolerability, feasibility, dose-limiting toxicity and maximum tolerated dose of topical NTG in addition to 5-fluorouracil and radiation therapy for neoadjuvant treatment of locoregionally advanced operable rectal cancer. The secondary endpoint was rate of pathologic complete response (pCR). Patients were assigned to 3 sequential cohorts of escalating dose levels of commercially available NTG patches (0.2, 0.4, and 0.6 mg/h), each cohort was intended to consist of 3 patients. RESULTS Thirteen patients were enrolled in the trial as specified in the dose escalation protocol. They were all male with a median age of 59.4 ± 2.5 (SEM) years. The observed toxicities were mild to moderate and manageable. Four patients developed asymptomatic grade 3 lymphopenia during the chemoradiation that resolved promptly upon completion. One patient had a non-ST segment elevation MI and 1 patient developed diarrhea. None of these toxicities were attributed to NTG except for 1 patient who developed a grade 3 headache. This required an additional group of patients at the same dose and no other patient experienced headaches. pCR was 17%. CONCLUSION NTG patches are well-tolerated and it is feasible to proceed with a phase II trial at the maximum dose examined (0.6 mg/h).
Collapse
Affiliation(s)
- Henrik Illum
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - David H Wang
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Jonathan E Dowell
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - William J Hittson
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - John R Torrisi
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Jeffrey Meyer
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX
| | - Sergio Huerta
- VA North Texas Health Care System Department of Hematology Oncology, Radiation Oncology, and Surgery/University of Texas Southwestern, Department of Surgery and Radiation Oncology, Dallas, TX.
| |
Collapse
|
17
|
Fulda S. Targeting IAP proteins in combination with radiotherapy. Radiat Oncol 2015; 10:105. [PMID: 25927408 PMCID: PMC4436972 DOI: 10.1186/s13014-015-0399-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 04/01/2015] [Indexed: 01/30/2023] Open
Abstract
The efficacy of radiotherapy critically depends on the activation of intrinsic cell death programs in cancer cells. This implies that evasion of cell death, a hallmark of human cancers, can contribute to radioresistance. Therefore, novel strategies to reactivate cell death programs in cancer cells are required in order to overcome resistance to radiotherapy. Since Inhibitor of Apoptosis (IAP) proteins are expressed at high levels in multiple cancers and block cell death induction at a central point, therapeutic targeting of IAP proteins represents a promising approach to potentiate the efficacy of radiotherapy. The current review discusses the concept of targeting IAP proteins in combination with radiotherapy.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528, Frankfurt, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
18
|
Lu J, Qin Q, Zhan LL, Liu J, Zhu HC, Yang X, Zhang C, Xu LP, Liu ZM, Wang D, Cui HQ, Meng CC, Cai J, Cheng HY, Sun XC. AT-406, an IAP inhibitor, activates apoptosis and induces radiosensitization of normoxic and hypoxic cervical cancer cells. J Pharmacol Sci 2014; 126:56-65. [PMID: 25168470 DOI: 10.1254/jphs.14079fp] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
IAP antagonists increased the antitumor efficacy of X-irradiation in some types of cancers, but their effects on hypoxic cancer cells remain unclarified. We aims to investigate the radiosensitizing effect of an IAP inhibitor AT-406 on cervical cancer cell lines under both normoxia and hypoxia conditions. Hela and Siha cells were treated to investigate the effects of drug administration on cell proliferation, apoptosis, and radiosensitivity. Western blot analysis was used to determine the role of AT-406 in inhibition of IAPs. The pathway of apoptosis was characterized by caspases activity assay. AT-406 potently sensitized Hela cells but not Siha cells to radiation under normoxia. Notably, the radiosensitizing effect of AT-406 on hypoxic cells was more evident than on normoxic cells in both cell lines. Further mechanism studies by western blot showed that under normoxia AT-406 decreased the level of cIAP1 in Hela cells in a dose-dependent manner; while additional downregulation of XIAP expression was induced by AT-406 treatment under hypoxia in both cell lines. Finally, AT-406 works on both extrinsic death receptor and intrinsic mitochondrial apoptosis pathways to activate apoptosis. Totally, AT-406 acts as a strong radiosensitizer in human cervical cancer cells, especially in hypoxic condition.
Collapse
Affiliation(s)
- Jing Lu
- Department of Radiation Oncology, the First Affiliated Hospital of Nanjing Medical University, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ramzan Z, Nassri AB, Huerta S. Genotypic characteristics of resistant tumors to pre-operative ionizing radiation in rectal cancer. World J Gastrointest Oncol 2014; 6:194-210. [PMID: 25024812 PMCID: PMC4092337 DOI: 10.4251/wjgo.v6.i7.194] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/19/2014] [Accepted: 05/08/2014] [Indexed: 02/05/2023] Open
Abstract
Due to a wide range of clinical response in patients undergoing neo-adjuvant chemoradiation for rectal cancer it is essential to understand molecular factors that lead to the broad response observed in patients receiving the same form of treatment. Despite extensive research in this field, the exact mechanisms still remain elusive. Data raging from DNA-repair to specific molecules leading to cell survival as well as resistance to apoptosis have been investigated. Individually, or in combination, there is no single pathway that has become clinically applicable to date. In the following review, we describe the current status of various pathways that might lead to resistance to the therapeutic applications of ionizing radiation in rectal cancer.
Collapse
|
20
|
Pouliliou S, Koukourakis MI. Gamma histone 2AX (γ-H2AX)as a predictive tool in radiation oncology. Biomarkers 2014; 19:167-80. [DOI: 10.3109/1354750x.2014.898099] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Stamatia Pouliliou
- Department of Radiotherapy/Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace
AlexandroupolisGreece
| | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace
AlexandroupolisGreece
| |
Collapse
|
21
|
Koehler BC, Jäger D, Schulze-Bergkamen H. Targeting cell death signaling in colorectal cancer: Current strategies and future perspectives. World J Gastroenterol 2014; 20:1923-1934. [PMID: 24587670 PMCID: PMC3934462 DOI: 10.3748/wjg.v20.i8.1923] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/06/2013] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
The evasion from controlled cell death induction has been considered as one of the hallmarks of cancer cells. Defects in cell death signaling are a fundamental phenomenon in colorectal cancer. Nearly any non-invasive cancer treatment finally aims to induce cell death. However, apoptosis resistance is the major cause for insufficient therapeutic success and disease relapse in gastrointestinal oncology. Various compounds have been developed and evaluated with the aim to meet with this obstacle by triggering cell death in cancer cells. The aim of this review is to illustrate current approaches and future directions in targeting cell death signaling in colorectal cancer. The complex signaling network of apoptosis will be demonstrated and the “druggability” of targets will be identified. In detail, proteins regulating mitochondrial cell death in colorectal cancer, such as Bcl-2 and survivin, will be discussed with respect to potential therapeutic exploitation. Death receptor signaling and targeting in colorectal cancer will be outlined. Encouraging clinical trials including cell death based targeted therapies for colorectal cancer are under way and will be demonstrated. Our conceptual understanding of cell death in cancer is rapidly emerging and new types of controlled cellular death have been identified. To meet this progress in cell death research, the implication of autophagy and necroptosis for colorectal carcinogenesis and therapeutic approaches will also be depicted. The main focus of this topic highlight will be on the revelation of the complex cell death concepts in colorectal cancer and the bridging from basic research to clinical use.
Collapse
|
22
|
Smac mimetic compound LCL161 sensitizes esophageal carcinoma cells to radiotherapy by inhibiting the expression of inhibitor of apoptosis protein. Tumour Biol 2013; 35:2565-74. [PMID: 24170321 DOI: 10.1007/s13277-013-1338-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/16/2013] [Indexed: 10/26/2022] Open
Abstract
Currently, unresectable esophageal squamous cell carcinoma (ESCC) is primarily treated by chemoradiotherapy. However, the outcome has not improved significantly due to radioresistance of cancer cells. This study aimed to determine the radiosensitizing effect of LCL161, a novel second mitochondrial-derived activator of caspase (Smac) mimetic, in ESCC cells. ESCC cell lines were treated with LCL161 or radiation, alone or in combination. Cell proliferation was detected by MTT assay. Radiosensitization was evaluated by clonogenic survival assay. Cell apoptosis was detected by flow cytometry. The results showed that LCL161 potently sensitized ESCC cells to radiation with a sensitization enhancement ratio of 1.4-2.0. LCL161 increased radiation-induced DNA double-stranded breaks and promoted the apoptosis of ESCC cells, which could be abrogated by a pan-caspase inhibitor z-VAD-FMK. Furthermore, LCL161 decreased the level of cIAP1 in ESCC cells in a dose-dependent manner and synthesized with irradiation to promote the activation of caspase 8 and the upregulation of TNFα expression in ESCC cells. In conclusion, LCL161 acts as a strong radiosensitizer in human esophageal cancer cells by inhibiting the expression of cIAP1 and promoting the activation of caspase 8, leading to enhanced apoptosis.
Collapse
|
23
|
Danquah M, Duke CB, Patil R, Miller DD, Mahato RI. Combination therapy of antiandrogen and XIAP inhibitor for treating advanced prostate cancer. Pharm Res 2012; 29:2079-91. [PMID: 22451249 DOI: 10.1007/s11095-012-0737-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Accepted: 03/09/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Overexpression of the androgen receptor (AR) and anti-apoptotic genes including X-linked inhibitor of apoptosis protein (XIAP) provide tumors with a proliferative advantage. Therefore, our objective was to determine whether novel antiandrogen (CBDIV17) and XIAP inhibitor based combination therapy can treat advanced prostate cancer. METHODS CBDIV17 and embelin-6g were synthesized and their effect on cell proliferation, apoptosis, cell cycle and AR and XIAP gene silencing determined. RESULTS CBDIV17 was more potent than bicalutamide and inhibited proliferation of C4-2 and LNCaP cells, IC(50) for CBDIV17 was ≈ 12 μM and ≈ 21 μM in LNCaP and C4-2 cells, respectively, whereas bicalutamide had IC(50) of ≈ 46 μM in LNCaP cells and minimal effect in C4-2 cells. CBDIV17 induced apoptosis more effectively compared to bicalutamide and significantly inhibited DNA replication. Combination of CBDIV17 and embelin resulted in supra-additive antiproliferative and apoptotic effects. Embelin downregulated AR expression and decreased androgen-mediated AR phosphorylation at Ser(81). These hydrophobic drugs were solubilized using micelles prepared with polyethylene glycol-b-poly (carbonate-co-lactide) (PEG-b-p(CB-co-LA)) copolymer. Combination therapy inhibited prostate tumor growth more effectively compared to control or monotherapy in vivo. CONCLUSIONS Our results demonstrated that CBDIV17 in combination with embelin can potentially treat advanced prostate cancer.
Collapse
Affiliation(s)
- Michael Danquah
- University of Tennessee Health Science Center, Department of Pharmaceutical Sciences, 19 South Manassas, CRB RM 226, Memphis, 38103-3308, Tennessee, USA
| | | | | | | | | |
Collapse
|
24
|
Inhibitor of Apoptosis (IAP) proteins as therapeutic targets for radiosensitization of human cancers. Cancer Treat Rev 2012; 38:760-6. [PMID: 22342104 DOI: 10.1016/j.ctrv.2012.01.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 01/17/2012] [Accepted: 01/23/2012] [Indexed: 01/22/2023]
Abstract
Radiotherapy initiates a variety of signaling events in cancer cells that eventually lead to cell death in case the DNA damage cannot be repaired. However, the signal transduction pathways that mediate cell death in response to radiation-inflicted DNA damage are frequently disturbed in human cancers, contributing to radioresistance. For example, aberrant activation of antiapoptotic programs such as high expression of Inhibitor of Apoptosis (IAP) proteins has been shown to interfere with the efficacy of radiotherapy. Since IAP proteins have been linked to radioresistance in several malignancies, therapeutic targeting of IAP proteins may open new perspectives to overcome radioresistance. Therefore, molecular targeting of IAP proteins may provide novel opportunities to reactivate cell death pathways that mediate radiation-induced cytotoxicity. A number of strategies have been developed in recent years to antagonize IAP proteins for the treatment of cancers. Some of these approaches have already been translated into a clinical application. While IAP protein-targeting agents are currently being evaluated in early clinical trials alone or in combination with conventional chemotherapy, they have not yet been tested in combination with radiation therapy. Therefore, it is a timely subject to discuss the opportunities of antagonizing IAP proteins for radiosensitization. Preclinical studies demonstrating the potential of this concept in relevant in vitro and in vivo models underscore that this combination approach warrants further clinical investigation. Thus, combination protocols using IAP antagonists together with radiotherapy may pave the avenue to more effective radiation-based treatment options for cancer patients.
Collapse
|
25
|
BV6, an IAP antagonist, activates apoptosis and enhances radiosensitization of non-small cell lung carcinoma in vitro. J Thorac Oncol 2012; 6:1801-9. [PMID: 21760551 DOI: 10.1097/jto.0b013e318226b4a6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Defects in the apoptosis pathway limit the effectiveness of radiation in non-small cell lung cancer (NSCLC) therapy. BV6 is an antagonist of cIAP1 and XIAP, members of the inhibitors of apoptosis (IAP) family. We investigated the potential of BV6 to sensitize NSCLC cell lines to radiation. METHODS HCC193 and H460 lung cancer cell lines were treated with BV6 to investigate the effects of drug administration on cell proliferation, apoptosis, inhibition of XIAP and cIAP1, and radiosensitivity. Subsequent immunoblotting and Hoechst staining were used to determine the role of apoptosis in radiosensitization. Finally, the pathway of apoptosis was characterized by Western blot analysis for cleaved caspase-8 and cleaved caspase-9 and enzyme-linked immunosorbent assays for TNF-α. RESULTS HCC193 was found to be more sensitive than H460 to BV6-induced apoptosis in a concentration-dependent and time-dependent manner. BV6 significantly sensitized both cell lines to radiation (HCC193-DER = 1.38, p < 0.05 at 1 μM BV6; H460-DER = 1.42, p < 0.05 at 5 μM BV6), but a higher concentration of and longer incubation time with BV6 was necessary for H460 cells. The BV6-induced radiosensitization of HCC193 favored the extrinsic pathway of apoptosis, whereas that of H460 favored the intrinsic pathway. CONCLUSIONS BV6, an IAP antagonist, significantly enhanced the radiosensitization of HCC193 and H460 cells in vitro. More research is warranted to test the mechanism of action of BV6 and to assess its potential in vivo and in the clinical setting.
Collapse
|
26
|
SASAKI KAZUHITO, TSUNO NELSONH, SUNAMI EIJI, KAWAI KAZUSHIGE, SHUNO YASUTAKA, HONGO KUMIKO, HIYOSHI MASAYA, KANEKO MANABU, MURONO KOJI, TADA NORIKO, NIREI TAKAKO, KITAYAMA JOJI, TAKAHASHI KOKI, NAGAWA HIROKAZU. Radiosensitization of human breast cancer cells to ultraviolet light by 5-fluorouracil. Oncol Lett 2011; 2:471-476. [PMID: 22866105 PMCID: PMC3410487 DOI: 10.3892/ol.2011.261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 02/22/2011] [Indexed: 11/05/2022] Open
Abstract
Ultraviolet light B (UVB) phototherapy is widely used to treat dermatological diseases and therefore may be a potential optional strategy in the treatment of a skin lesion infiltrated by a malignant tumor. Currently, little is known regarding the effect of UVB phototherapy on human breast cancer cells. The present study aimed to investigate the effect of UVB phototherapy, as well as the potential effect of 5-fluorouracil (5-FU), the first-line anticancer drug for breast cancer, on radiosensitizing MCF-7 human breast cancer cells, in an attempt to develop new therapeutic strategies for the treatment of locoregional recurrence of breast cancer. MCF-7 cells were incubated in the presence of 5-FU for 48 h, and UVB irradiation at 750 mJ/cm(2) was administered in the midterm of 5-FU treatment. The viability of MCF-7 cells was analyzed by the trypan blue staining method. Apoptosis was quantified by flow cytometry and Hoechst 33258 staining. The cell cycle was evaluated by flow cytometry after the staining of cells with propidium iodide. The combination treatment of 5-FU and UVB resulted in a strong potentiation of the inhibitory effect of MCF-7 cell growth, dependent on the intra-S phase cell cycle arrest and induction of apoptosis, when compared to treatment with 5-FU or UVB alone. In conclusion, 5-FU sensitized human breast cancer cells to UVB phototherapy, and this combination therapy is an effective and promising strategy for the treatment of breast cancer, particularly for locoregional recurrence.
Collapse
Affiliation(s)
- KAZUHITO SASAKI
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - NELSON H. TSUNO
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
- Department of Transfusion Medicine, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - EIJI SUNAMI
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - KAZUSHIGE KAWAI
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - YASUTAKA SHUNO
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - KUMIKO HONGO
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - MASAYA HIYOSHI
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - MANABU KANEKO
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - KOJI MURONO
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - NORIKO TADA
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - TAKAKO NIREI
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - JOJI KITAYAMA
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - KOKI TAKAHASHI
- Department of Transfusion Medicine, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| | - HIROKAZU NAGAWA
- Department of Surgical Oncology, Faculty of Medical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
27
|
Cheung HH, Beug ST, St Jean M, Brewster A, Kelly NL, Wang S, Korneluk RG. Smac mimetic compounds potentiate interleukin-1beta-mediated cell death. J Biol Chem 2010; 285:40612-23. [PMID: 20956527 DOI: 10.1074/jbc.m110.183616] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Smac mimetic compounds (SMCs) potentiate TNFα-mediated cancer cell death by targeting the inhibitor of apoptosis (IAP) proteins. In addition to TNFα, the tumor microenvironment is exposed to a number of pro-inflammatory cytokines, including IL-1β. Here, we investigated the potential impact of IL-1β on SMC-mediated death of cancer cells. Synergy was seen in a subset of a diverse panel of 21 cancer cell lines to the combination of SMC and IL-1β treatment, which required IL-1β-induced activation of the NF-κB pathway. Elevated NF-κB activity resulted in the production of TNFα, which led to apoptosis dependent on caspase-8 and RIP1. In addition, concurrent silencing of cIAP1, cIAP2, and X-linked IAP by siRNA was most effective for triggering IL-1β-mediated cell death. Importantly, SMC-resistant cells that produced TNFα in response to IL-1β treatment were converted to an SMC-sensitive phenotype by c-FLIP knockdown. Reciprocally, ectopic expression of c-FLIP blocked cell death caused by combined SMC and IL-1β treatment in sensitive cancer cells. Together, our study indicates that a positive feed-forward loop by pro-inflammatory cytokines can be exploited by SMCs to induce apoptosis in cancer cells.
Collapse
Affiliation(s)
- Herman H Cheung
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, Ottawa, Ontario K1H 8L1, Canada
| | | | | | | | | | | | | |
Collapse
|