1
|
Saurav S, Karfa S, Vu T, Liu Z, Datta A, Manne U, Samuel T, Datta PK. Overcoming Irinotecan Resistance by Targeting Its Downstream Signaling Pathways in Colon Cancer. Cancers (Basel) 2024; 16:3491. [PMID: 39456585 PMCID: PMC11505920 DOI: 10.3390/cancers16203491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Among the most popular chemotherapeutic agents, irinotecan, regarded as a prodrug belonging to the camptothecin family that inhibits topoisomerase I, is widely used to treat metastatic colorectal cancer (CRC). Although immunotherapy is promising for several cancer types, only microsatellite-instable (~7%) and not microsatellite-stable CRCs are responsive to it. Therefore, it is important to investigate the mechanism of irinotecan function to identify cellular proteins and/or pathways that could be targeted for combination therapy. Here, we have determined the effect of irinotecan treatment on the expression/activation of tumor suppressor genes (including p15Ink4b, p21Cip1, p27Kip1, and p53) and oncogenes (including OPN, IL8, PD-L1, NF-κB, ISG15, Cyclin D1, and c-Myc) using qRT-PCR, Western blotting, immunofluorescence (IF), and RNA sequencing of tumor specimens. We employed stable knockdown, neutralizing antibodies (Abs), and inhibitors of OPN, p53, and NF-κB to establish downstream signaling and sensitivity/resistance to the cytotoxic activities of irinotecan. Suppression of secretory OPN and NF-κB sensitized colon cancer cells to irinotecan. p53 inhibition or knockdown was not sufficient to block or potentiate SN38-regulated signaling, suggesting p53-independent effects. Irinotecan treatment inhibited tumor growth in syngeneic mice. Analyses of allograft tumors from irinotecan-treated mice validated the cell culture results. RNA-seq data suggested that irinotecan-mediated activation of NF-κB signaling modulated immune and inflammatory genes in mice, which may compromise drug efficacy and promote resistance. In sum, these results suggest that, for CRCs, targeting OPN, NF-κB, PD-L1, and/or ISG15 signaling may provide a potential strategy to overcome resistance to irinotecan-based chemotherapy.
Collapse
Affiliation(s)
- Shashank Saurav
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Sourajeet Karfa
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Trung Vu
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| | - Zhipeng Liu
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Arunima Datta
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Temesgen Samuel
- Department of Pathobiology, Tuskegee University, Tuskegee, AL 36088, USA
| | - Pran K. Datta
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
- Birmingham Veterans Affairs Medical Center, Birmingham, AL 35233, USA
| |
Collapse
|
2
|
Gomez Rodriguez Y, Oliva Arguelles B, Riera-Romo M, Fernandez-De-Cossio J, Garay HE, Fernandez Masso J, Guerra Vallespi M. Synergic effect of anticancer peptide CIGB-552 and Cisplatin in lung cancer models. Mol Biol Rep 2022; 49:3197-3212. [DOI: 10.1007/s11033-022-07152-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 01/17/2022] [Indexed: 12/24/2022]
|
3
|
He H, Shao X, Li Y, Gihu R, Xie H, Zhou J, Yan H. Targeting Signaling Pathway Networks in Several Malignant Tumors: Progresses and Challenges. Front Pharmacol 2021; 12:675675. [PMID: 34135756 PMCID: PMC8203325 DOI: 10.3389/fphar.2021.675675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 05/18/2021] [Indexed: 12/22/2022] Open
Abstract
Malignant tumors remain the health problem of highest concern among people worldwide due to its high mortality and recurrence. Lung, gastric, liver, colon, and breast cancers are among the top five malignant tumors in terms of morbidity and mortality. In cancer biology, aberrant signaling pathway regulation is a prevalent theme that drives the generation, metastasis, invasion, and other processes of all malignant tumors. The Wnt/β-catenin, PI3K/AKT/mTOR, Notch and NF-kB pathways are widely concerned and signal crosstalks exist in the five solid tumors. This review provides an innovative summary of the recent progress in research on these signaling pathways, the underlying mechanism of the molecules involved in these pathways, and the important role of some miRNAs in tumor-related signaling pathways. It also presents a brief review of the antitumor molecular drugs that target these signaling pathways. This review may provide a theoretical basis for the study of the molecular biological mechanism of malignant tumors and vital information for the development of new treatment strategies with a focus on efficacy and the reduction of side effects.
Collapse
Affiliation(s)
- Hongdan He
- Qinghai Tibet Plateau Research Institute, Southwest Minzu University, Chengdu, China
| | - Xiaoni Shao
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Yanan Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Ribu Gihu
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Haochen Xie
- Qinghai Tibet Plateau Research Institute, Southwest Minzu University, Chengdu, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
4
|
Transcription factors in colorectal cancer: molecular mechanism and therapeutic implications. Oncogene 2020; 40:1555-1569. [PMID: 33323976 DOI: 10.1038/s41388-020-01587-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a major cause of cancer mortality worldwide, however, the molecular mechanisms underlying the pathogenesis of CRC remain largely unclear. Recent studies have revealed crucial roles of transcription factors in CRC development. Transcription factors essential for the regulation of gene expression by interacting with transcription corepressor/enhancer complexes and they orchestrate downstream signal transduction. Deregulation of transcription factors is a frequent occurrence in CRC, and the accompanying drastic changes in gene expression profiles play fundamental roles in multistep process of tumorigenesis, from cellular transformation, disease progression to metastatic disease. Herein, we summarized current and emerging key transcription factors that participate in CRC tumorigenesis, and highlighted their oncogenic or tumor suppressive functions. Moreover, we presented critical transcription factors of CRC, emphasized the major molecular mechanisms underlying their effect on signal cascades associated with tumorigenesis, and summarized of their potential as molecular biomarkers for CRC prognosis therapeutic response, as well as drug targets for CRC treatment. A better understanding of transcription factors involved in the development of CRC will provide new insights into the pathological mechanisms and reveal novel prognostic biomarkers and therapeutic strategies for CRC.
Collapse
|
5
|
Sadaghianloo N, Contenti J, Dufies M, Parola J, Rouleau M, Lee S, Peyron J, Fabbri L, Hassen‐Khodja R, Pouysségur J, Bost F, Jean‐Baptiste E, Dardik A, Mazure NM. Co-culture of human fibroblasts, smooth muscle and endothelial cells promotes osteopontin induction in hypoxia. J Cell Mol Med 2020; 24:2931-2941. [PMID: 32032472 PMCID: PMC7077551 DOI: 10.1111/jcmm.14905] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/20/2019] [Accepted: 11/23/2019] [Indexed: 12/17/2022] Open
Abstract
Arteriovenous fistulas (AVFs) are the preferred vascular access for haemodialysis of patients suffering from end-stage renal disease, a worldwide public health problem. However, they are prone to a high rate of failure due to neointimal hyperplasia and stenosis. This study aimed to determine if osteopontin (OPN) was induced in hypoxia and if OPN could be responsible for driving AVF failure. Identification of new factors that participate in remodelling of AVFs is a challenge. Three cell lines representing the cells of the three layers of the walls of arteries and veins, fibroblasts, smooth muscle cells and endothelial cells, were tested in mono- and co-culture in vitro for OPN expression and secretion in normoxia compared to hypoxia after silencing the hypoxia-inducible factors (HIF-1α, HIF-2α and HIF-1/2α) with siRNA or after treatment with an inhibitor of NF-kB. None of the cells in mono-culture showed OPN induction in hypoxia, whereas cells in co-culture secreted OPN in hypoxia. The changes in oxygenation that occur during AVF maturation up-regulate secretion of OPN through cell-cell interactions between the different cell layers that form AVF, and in turn, these promote endothelial cell proliferation and could participate in neointimal hyperplasia.
Collapse
Affiliation(s)
- Nirvana Sadaghianloo
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Julie Contenti
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Department of Emergency MedicineCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | | | - Julien Parola
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | | | - Shinrong Lee
- Department of Surgery and the Vascular Biology and Therapeutics ProgramYale UniversityNew HavenCTUSA
- Department of Vascular SurgeryVA Connecticut Healthcare SystemsWest HavenCTUSA
| | - Jean‐François Peyron
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | - Lucilla Fabbri
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Réda Hassen‐Khodja
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Jacques Pouysségur
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Centre Scientifique de Monaco (CSM)MonacoMonaco
| | - Frédéric Bost
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
| | - Elixène Jean‐Baptiste
- Department of Vascular SurgeryCentre Hospitalier Universitaire de NiceNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| | - Alan Dardik
- Department of Surgery and the Vascular Biology and Therapeutics ProgramYale UniversityNew HavenCTUSA
- Department of Vascular SurgeryVA Connecticut Healthcare SystemsWest HavenCTUSA
| | - Nathalie M. Mazure
- Université Côte d’AzurInstitute for Research on Cancer and Aging of Nice (IRCAN)CNRS‐UMR 7284‐Inserm U1081Centre Antoine LacassagneUniversity of Nice Sophia‐AntipolisNiceFrance
- Present address:
Centre de Méditerranéen de Médecine Moléculaire (C3M)INSERM U1065Université Côte d’AzurNice Cedex 03France
| |
Collapse
|
6
|
Soleimani A, Rahmani F, Ferns GA, Ryzhikov M, Avan A, Hassanian SM. Role of the NF-κB signaling pathway in the pathogenesis of colorectal cancer. Gene 2019; 726:144132. [PMID: 31669643 DOI: 10.1016/j.gene.2019.144132] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 09/12/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
The NF-κB signaling pathway is a key regulator of CRC cell proliferation, apoptosis, angiogenesis, inflammation, metastasis, and drug resistance. Over-activation of the NF-κB pathway is a feature of colorectal cancer (CRC). While new combinatorial treatments have improved overall patient outcome; quality of life, cost of care, and patient survival rate have seen little improvement. Suppression of the NF-κB signaling pathway using biological or specific pharmacological inhibitors is a potential therapeutic approach in the treatment of colon cancer. This review summarizes the regulatory role of NF-κB signaling pathway in the pathogenesis of CRC for a better understanding and hence a better management of the disease.
Collapse
Affiliation(s)
- Atena Soleimani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Mikhail Ryzhikov
- Division of Pulmonary and Critical Care Medicine, Washington University, School of Medicine, Saint Louis, MO, USA
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Zhou X, Shan Z, Yang H, Xu J, Li W, Guo F. RelB plays an oncogenic role and conveys chemo-resistance to DLD-1 colon cancer cells. Cancer Cell Int 2018; 18:181. [PMID: 30473630 PMCID: PMC6234565 DOI: 10.1186/s12935-018-0677-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/04/2018] [Indexed: 12/24/2022] Open
Abstract
Background Nuclear transcription factor kappa B (NF-κB) subunits exhibit crucial roles in tumorigenesis and chemo-sensitivity. Recent studies suggest that RelB, the key subunit of the alternative NF-κB pathway, plays a critical role in the progression of diverse human malignancies. However, the significance of RelB in colorectal cancer (CRC) remains unclear. Here, we systematically explored the functions of the alternative NF-κB subunit RelB in colon cancer cells and its underlying mechanism. Methods Stably transfected RelB-shRNA DLD-1 cells were established using Lipofectamine 2000. NF-κB DNA-binding capability was quantified using an ELISA-based NF-κB activity assay. Cell growth was monitored by an x-Celligence system. Cell proliferation was analyzed by a CCK-8 and a Brdu proliferation assay. Response to 5-FU was assessed by an x-Celligence system. Cell apoptosis and cell cycle was detected using flow cytometry analyses. Cell migration and invasion abilities were detected by an x-Celligence system, Transwell inserts, and wound-healing assays. RelB expression and its clinical significance were analyzed using the CRC tissue microarray. The expression of NF-κB signaling subunits, AKT/mTOR signaling molecules, cell cycle related proteins, MMP2, MMP9, and Integrin β-1 were measured by Western blotting analyses. Results The RelB-silencing inhibited cell growth of DLD-1 cells. The RelB-silencing exerted the anti-proliferative by downregulation of AKT/mTOR signaling. The RelB-silencing caused G0–G1 cell cycle arrested likely due to decreasing the expression of Cyclin D1 and CDK4, concomitant with increased expression of p27Kip1. The RelB-silencing enhanced cytotoxic effect of 5-FU and induced cell accumulation in S-phase. The RelB-silencing impaired the migration and invasion potential of DLD-1 cells, which was related to downregulation of MMP2, MMP9, and Integrin β-1. Importantly, the RelB expression was correlated with depth of tumor invasion, lymph node metastasis, metastasis stage, and pTNM stage. High-RelB expression was significantly correlated with poor overall survival in CRC patients. Conclusion Our studies here provided evidence that RelB plays an oncogenic role and conveys chemo-resistance to 5-FU. RelB can be considered as an independent indicator of prognosis in CRC.
Collapse
Affiliation(s)
- Xiaojun Zhou
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Zhili Shan
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Hengying Yang
- 1Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Jingjing Xu
- 2Center for Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, 215006 China
| | - Wenjing Li
- 3Department of Clinical Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215006 China
| | - Feng Guo
- 4Department of Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Baita West Road 16, Suzhou, 215001 China
| |
Collapse
|
8
|
NF-κB pathways in the development and progression of colorectal cancer. Transl Res 2018; 197:43-56. [PMID: 29550444 DOI: 10.1016/j.trsl.2018.02.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/12/2022]
Abstract
Nuclear factor-κB (NF-κB) has been widely implicated in the development and progression of cancer. In colorectal cancer (CRC), NF-κB has a key role in cancer-related processes such as cell proliferation, apoptosis, angiogenesis, and metastasis. The role of NF-κB in CRC is complex, owed to the cross talk with other signaling pathways. Although there is sufficient evidence gained from cell lines and animal models that NF-κB is involved in cancer-related processes, because of a lack of studies in human tissue, the clinical evidence of its importance is limited in patients with CRC. This review summarizes evidence relating to how NF-κB is involved in the development and progression of CRC and comments on future work to be carried out.
Collapse
|
9
|
Bharti AC, Rajan P, Jadli M, Pande D, Singh T, Bhat A. Berberine as an Adjuvant and Sensitizer to Current Chemotherapy. ROLE OF NUTRACEUTICALS IN CHEMORESISTANCE TO CANCER 2018:221-240. [DOI: 10.1016/b978-0-12-812373-7.00011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Oleanolic Acid Alters Multiple Cell Signaling Pathways: Implication in Cancer Prevention and Therapy. Int J Mol Sci 2017; 18:ijms18030643. [PMID: 28300756 PMCID: PMC5372655 DOI: 10.3390/ijms18030643] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 12/13/2022] Open
Abstract
Nowadays, much attention has been paid to diet and dietary supplements as a cost-effective therapeutic strategy for prevention and treatment of a myriad of chronic and degenerative diseases. Rapidly accumulating scientific evidence achieved through high-throughput technologies has greatly expanded the understanding about the multifaceted nature of cancer. Increasingly, it is being realized that deregulation of spatio-temporally controlled intracellular signaling cascades plays a contributory role in the onset and progression of cancer. Therefore, targeting regulators of oncogenic signaling cascades is essential to prevent and treat cancer. A plethora of preclinical and epidemiological evidences showed promising role of phytochemicals against several types of cancer. Oleanolic acid, a common pentacyclic triterpenoid, is mainly found in olive oil, as well as several plant species. It is a potent inhibitor of cellular inflammatory process and a well-known inducer of phase 2 xenobiotic biotransformation enzymes. Main molecular mechanisms underlying anticancer effects of oleanolic acid are mediated by caspases, 5' adenosine monophosphate-activated protein kinase, extracellular signal-regulated kinase 1/2, matrix metalloproteinases, pro-apoptotic Bax and bid, phosphatidylinositide 3-kinase/Akt1/mechanistic target of rapamycin, reactive oxygen species/apoptosis signal-regulating kinase 1/p38 mitogen-activated protein kinase, nuclear factor-κB, cluster of differentiation 1, CKD4, s6k, signal transducer and activator of transcription 3, as well as aforementioned signaling pathways . In this work, we critically review the scientific literature on the molecular targets of oleanolic acid implicated in the prevention and treatment of several types of cancer. We also discuss chemical aspects, natural sources, bioavailability, and safety of this bioactive phytochemical.
Collapse
|
11
|
Gao L, Xu Z, Wang Y, Sun B, Song Z, Yang B, Liu X, Lin Y, Peng J, Han G, Wang S, Tang Z. Anticancer effect of SZC017, a novel derivative of oleanolic acid, on human gastric cancer cells. Oncol Rep 2016; 35:1101-1108. [PMID: 26718492 DOI: 10.3892/or.2015.4447] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/06/2015] [Indexed: 11/05/2022] Open
Abstract
Oleanolic acid (OA) and its several derivatives possess chemopreventive and chemotherapeutic functions against a series of cancer types. Many chemotherapeutic compounds are effective in improving the quality of life and prolonging the survival of patients with gastric cancer, therefore progress in the treatment of gastric cancer, especially the anticancer effects of OA derivatives must be achieved. The inhibitory effect of SZC017, a newly synthesized derivative of OA, on cell viability was determined by MTT assay. Furthermore, flow cytometry, transmission electron microscopy, and western blot analysis revealed that the inhibition of cell viability by OA was mediated by triggering the intrinsic apoptosis of gastric cancer cells, and inducing S phase arrest of SGC7901 cells. Mechanistically, SZC017 was effective against gastric cancer cells via inhibiting Akt/NF‑κB signaling and topoisomerase I and IIα proteins. Taken together, our data indicate that SZC017 may be a potential chemotherapeutic agent against gastric cancer cells.
Collapse
Affiliation(s)
- Lei Gao
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhen Xu
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yan Wang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Bin Sun
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Zhicheng Song
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, P.R. China
| | - Bining Yang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Xu Liu
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yuan Lin
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jinyong Peng
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Guozhu Han
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Shisheng Wang
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, Liaoning 116024, P.R. China
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
12
|
Zhang SY, Cao N, Chen MK, Zhang LD, Bai YQ, Yang YX. Diagnostic value of plasma HLA-F adjacent transcript 10 mRNA expression in patients with hepatocellular carcinoma or colon cancer. Shijie Huaren Xiaohua Zazhi 2015; 23:5775-5783. [DOI: 10.11569/wcjd.v23.i36.5775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To detect plasma levels of human leukocyte antigen (HLA)-F adjacent transcript 10 (FAT10) mRNA in patients with hepatocellular carcinoma (HCC) or colon cancer (CC), and to assess its diagnostic value.
METHODS: Blood samples were collected from 70 patients with HCC, 60 patients with cirrhosis, 64 patients with CC, and 64 healthy volunteers. The expression levels of plasma FAT10 mRNA and α-fetoprotein (AFP) protein were measured by RT-PCR and ELISA, respectively.
RESULTS: The positive rates of FAT10 mRNA in plasma of patients with HCC, those with cirrhosis and healthy controls were 72.85%, 28.3% and 12.5%, respectively, and there were significantly differences in the three groups (P < 0.05). High expression levels of FAT10 mRNA in HCC were related to TNM stage and lymph node metastasis (P < 0.05). The positive rate of FAT10 mRNA was also significantly higher in CC patients than in healthy controls (P < 0.05). The up-regulation of FAT10 mRNA in CC was closely related to Duke's stage and distance metastasis. FAT10 mRNA had a sensitivity of 66.6% for diagnosis of AFP-negative HCC. Furthermore, the FAT10 mRNA had a sensitivity of 68.8% and specificity of 79.8% in distinguishing small HCC patients from non-HCC individuals (cirrhosis patients and healthy controls). Combined AFP and FAT10 mRNA detection could improve the sensitivity to 84.4%.
CONCLUSION: FAT10 mRNA is a promising serological marker for HCC and CC. Plasma FAT10 mRNA and AFP protein could be combined to improve the diagnosis of HCC.
Collapse
|
13
|
Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Arrojo R, Liu D, Rossi EA, Chang CH, Goldenberg DM. Sacituzumab Govitecan (IMMU-132), an Anti-Trop-2/SN-38 Antibody-Drug Conjugate: Characterization and Efficacy in Pancreatic, Gastric, and Other Cancers. Bioconjug Chem 2015; 26:919-31. [PMID: 25915780 DOI: 10.1021/acs.bioconjchem.5b00223] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Sacituzumab govitecan (IMMU-132) is an antibody-drug conjugate (ADC) made from a humanized anti-Trop-2 monoclonal antibody (hRS7) conjugated with the active metabolite of irinotecan, SN-38. In addition to its further characterization, as the clinical utility of IMMU-132 expands to an ever-widening range of Trop-2-expressing solid tumor types, its efficacy in new disease models needs to be explored in a nonclinical setting. Unlike most ADCs that use ultratoxic drugs and stable linkers, IMMU-132 uses a moderately toxic drug with a moderately stable carbonate bond between SN-38 and the linker. Flow cytometry and immunohistochemistry disclosed that Trop-2 is expressed in a wide range of tumor types, including gastric, pancreatic, triple-negative breast (TNBC), colonic, prostate, and lung. While cell-binding experiments reveal no significant differences between IMMU-132 and parental hRS7 antibody, surface plasmon resonance analysis using a Trop-2 CM5 chip shows a significant binding advantage for IMMU-132 over hRS7. The conjugate retained binding to the neonatal receptor, but it lost greater than 60% of the antibody-dependent cell-mediated cytotoxicity activity compared to that of hRS7. Exposure of tumor cells to either free SN-38 or IMMU-132 demonstrated the same signaling pathways, with pJNK1/2 and p21(WAF1/Cip1) upregulation followed by cleavage of caspases 9, 7, and 3, ultimately leading to poly-ADP-ribose polymerase cleavage and double-stranded DNA breaks. Pharmacokinetics of the intact ADC in mice reveals a mean residence time (MRT) of 15.4 h, while the carrier hRS7 antibody cleared at a similar rate as that of the unconjugated antibody (MRT ∼ 300 h). IMMU-132 treatment of mice bearing human gastric cancer xenografts (17.5 mg/kg; twice weekly × 4 weeks) resulted in significant antitumor effects compared to that of mice treated with a nonspecific control. Clinically relevant dosing schemes of IMMU-132 administered either every other week, weekly, or twice weekly in mice bearing human pancreatic or gastric cancer xenografts demonstrate similar, significant antitumor effects in both models. Current Phase I/II clinical trials ( ClinicalTrials.gov , NCT01631552) confirm anticancer activity of IMMU-132 in cancers expressing Trop-2, including gastric and pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Robert M Sharkey
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Preeti Trisal
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Roberto Arrojo
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Donglin Liu
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States
| | - Edmund A Rossi
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - Chien-Hsing Chang
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States
| | - David M Goldenberg
- †Immunomedics, Inc., Morris Plains, New Jersey 07950, United States.,‡IBC Pharmaceuticals, Inc., Morris Plains, New Jersey 07950, United States.,§Center of Molecular Medicine and Immunology, Garden State Cancer Center, Morris Plains, New Jersey 07950, United States
| |
Collapse
|
14
|
Yagishita S, Fujita Y, Kitazono S, Ko R, Nakadate Y, Sawada T, Kitamura Y, Shimoyama T, Maeda Y, Takahashi F, Takahashi K, Tamura T, Koizumi F. Chemotherapy-Regulated microRNA-125-HER2 Pathway as a Novel Therapeutic Target for Trastuzumab-Mediated Cellular Cytotoxicity in Small Cell Lung Cancer. Mol Cancer Ther 2015; 14:1414-23. [PMID: 25833836 DOI: 10.1158/1535-7163.mct-14-0625] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 03/10/2015] [Indexed: 11/16/2022]
Abstract
Small cell lung cancer (SCLC) accounts for 15% of all lung cancer cases and is a highly lethal disease. For the last several decades, the standard treatment for SCLC has been deadlocked, and new therapeutic strategies are urgently needed. HER2 is a member of the HER family and has been reported to be overexpressed in 30% of SCLC cases with poor prognosis. However, the clinical relevance of HER2-targeted therapy for SCLC remains unclear. Here, we first identify that cytotoxic drugs induce significant HER2 overexpression through microRNA-125a (miR-125a) and miR-125b downregulation, which in turn act as a novel therapeutic target for trastuzumab-mediated cellular cytotoxicity in SCLC. In this study, we showed that treatment of the HER2-positive SCLC cells, SBC-3 and SBC-5, with cytotoxic drugs induced a significant upregulation of HER2. Cisplatin (CDDP) treatment of SCLC cells resulted in a significant downregulation of miR-125a and miR-125b. We confirmed that miR-125a and miR-125b bound to the 3'-untranslated regions of HER2 mRNA, and that downregulation of miR-125a and miR-125b resulted in upregulation of HER2 in SCLC cells, suggesting a relationship between cytotoxic drug exposure and miR-125/HER2 dysregulation. Furthermore, using a calcein assay, we demonstrated a significantly enhanced cytotoxic effect of CDDP and trastuzumab that was mediated via antibody-dependent cellular cytotoxicity. Finally, we clearly demonstrated the enhanced antitumor effect of these agents in an orthotopic lung cancer model in vivo. Our results offer a novel therapeutic strategy for HER2-positive SCLCs by using trastuzumab combined with cytotoxic drugs.
Collapse
Affiliation(s)
- Shigehiro Yagishita
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan. Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yu Fujita
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tsukiji, Chuo-ku, Tokyo, Japan. Division of Respiratory Diseases, Department of Internal Medicine, Jikei University School of Medicine, Nishi-shinbashi, Minato-ku, Tokyo, Japan
| | - Satoru Kitazono
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan. Department of Thoracic Oncology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Ryo Ko
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Nakadate
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Takeshi Sawada
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Yuka Kitamura
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Tatsu Shimoyama
- Department of Chemotherapy, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
| | - Yoshiharu Maeda
- Department of Chemotherapy, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan
| | - Fumiyuki Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Kazuhisa Takahashi
- Department of Respiratory Medicine, Juntendo University Graduate School of Medicine, Hongo, Bunkyo-ku, Tokyo, Japan
| | - Tomohide Tamura
- Department of Thoracic Oncology, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan
| | - Fumiaki Koizumi
- Shien-Lab, National Cancer Center Hospital, Tsukiji, Chuo-ku, Tokyo, Japan. Department of Laboratory Medicine and Division of Clinical Research Support, Tokyo Metropolitan Cancer and Infectious Disease Center, Komagome Hospital, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
15
|
YU MEILING, TONG XUHUI, QI BENQUAN, QU HONGDANG, DONG SHUYING, YU BINBIN, ZHANG NAIJU, TANG NAN, WANG LINGZHI, ZHANG CUILING. Berberine enhances chemosensitivity to irinotecan in colon cancer via inhibition of NF-κB. Mol Med Rep 2013; 9:249-54. [DOI: 10.3892/mmr.2013.1762] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2013] [Accepted: 10/17/2013] [Indexed: 11/05/2022] Open
|
16
|
Fenouille N, Grosso S, Yunchao S, Mary D, Pontier-Bres R, Imbert V, Czerucka D, Caroli-Bosc FX, Peyron JF, Lagadec P. Calpain 2-dependent IκBα degradation mediates CPT-11 secondary resistance in colorectal cancer xenografts. J Pathol 2012; 227:118-29. [PMID: 22069124 DOI: 10.1002/path.3034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 09/20/2011] [Accepted: 10/21/2011] [Indexed: 12/12/2022]
Abstract
CPT-11 (irinotecan), the first-line chemotherapy for advanced stage colorectal cancer, remains inactive in about half of patients (primary chemoresistance) and almost all initial responders develop secondary resistance after several courses of treatment (8 months on average). Nude mice bearing HT-29 colon cancer xenografts were treated with CPT-11 and/or an NF-κB inhibitor for two courses. We confirm that NF-κB inhibition potentiated CPT-11 anti-tumoural effect after the first course of treatment. However, tumours grew again at the end of the second course of treatment, generating resistant tumours. We observed an increase in the basal NF-κB activation in resistant tumours and in two resistant sublines, either obtained from resistant HT-29 tumours (HT-29R cells) or generated in vitro (RSN cells). The decrease of NF-κB activation in HT-29R and RSN cells by stable transfections with the super-repressor form of IκBα augmented their sensitivity to CPT-11. Comparing gene expression profiles of HT-29 and HT-29R cells, we identified the S100A10/Annexin A2 complex and calpain 2 as over-expressed potential NF-κB inducers. SiRNA silencing of calpain 2 but not of S100A10 and/or annexin A2, resulted in a decrease in NF-κB activation, an increase in cellular levels of IκBα and a partial restoration of the CPT-11 sensitivity in both HT-29R and RSN cells, suggesting that calpain 2-dependent IκBα degradation mediates CPT-11 secondary resistance. Thus, targeted therapies directed against calpain 2 may represent a novel strategy to enhance the anti-cancer efficacy of CPT-11.
Collapse
Affiliation(s)
- Nina Fenouille
- INSERM, U895, Centre Méditerranéen de Médecine Moléculaire (C3M), Equipe Inflammation, Cancer, Cellules Souches Cancéreuses, Nice, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Jayanthan A, Bernoux D, Bose P, Riabowol K, Narendran A. Multi-tyrosine kinase inhibitors in preclinical studies for pediatric CNS AT/RT: Evidence for synergy with Topoisomerase-I inhibition. Cancer Cell Int 2011; 11:44. [PMID: 22206574 PMCID: PMC3278350 DOI: 10.1186/1475-2867-11-44] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 12/29/2011] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Currently, Atypical Teratoid Rhabdoid Tumor (AT/RT) constitutes one of the most difficult to treat malignancies in pediatrics. Hence, new knowledge of potential targets for therapeutics and the development of novel treatment approaches are urgently needed. We have evaluated the presence of cytokine pathways and the effects of two clinically available multi-tyrosine kinase inhibitors for cytotoxicity, target modulation and drug combinability against AT/RT cell lines. RESULTS AT/RT cell lines expressed measurable quantities of VEGF, FGF, PDGF and SDF-1, although the absolute amounts varied between the cell lines. The targeted receptor tyrosine kinase inhibitor sorafenib inhibited the key signaling molecule Erk, which was activated following the addition of own conditioned media, suggesting the existence of autocrine/paracrine growth stimulatory pathways. The multi-tyrosine kinase inhibitors sorafenib and sunitinib also showed significant growth inhibition of AT/RT cells and their activity was enhanced by combination with the topoisomerase inhibitor, irinotecan. The loss of cytoplasmic NF-kappa-B in response to irinotecan was diminished by sorafenib, providing evidence for a possible benefit for this drug combination. CONCLUSIONS In addition to previously described involvement of insulin like growth factor (IGF) family of cytokines, a multitude of other growth factors may contribute to the growth and survival of AT/RT cells. However, consistent with the heterogeneous nature of this tumor, quantitative and qualitative differences may exist among different tumor samples. Multi-tyrosine kinase inhibitors appear to have effective antitumor activity against all cell lines studied. In addition, the target modulation studies and drug combinability data provide the groundwork for additional studies and support the evaluation of these agents in future treatment protocols.
Collapse
Affiliation(s)
- Aarthi Jayanthan
- Laboratory for Pre-clinical and Drug Discovery Studies, Pediatric Oncology Experimental Therapeutics Investigators Consortium (POETIC) and Division of Pediatric Oncology, Alberta Children's Hospital, 2888 Shaganappi Trail NW, Calgary, T3B 6A8, Canada.
| | | | | | | | | |
Collapse
|
18
|
Cardillo TM, Govindan SV, Sharkey RM, Trisal P, Goldenberg DM. Humanized anti-Trop-2 IgG-SN-38 conjugate for effective treatment of diverse epithelial cancers: preclinical studies in human cancer xenograft models and monkeys. Clin Cancer Res 2011; 17:3157-69. [PMID: 21372224 PMCID: PMC10766325 DOI: 10.1158/1078-0432.ccr-10-2939] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Evaluate the efficacy of an SN-38-anti-Trop-2 antibody-drug conjugate (ADC) against several human solid tumor types, and to assess its tolerability in mice and monkeys, the latter with tissue cross-reactivity to hRS7 similar to humans. EXPERIMENTAL DESIGN Two SN-38 derivatives, CL2-SN-38 and CL2A-SN-38, were conjugated to the anti-Trop-2-humanized antibody, hRS7. The immunoconjugates were characterized in vitro for stability, binding, and cytotoxicity. Efficacy was tested in five different human solid tumor-xenograft models that expressed Trop-2 antigen. Toxicity was assessed in mice and in Cynomolgus monkeys. RESULTS The hRS7 conjugates of the two SN-38 derivatives were equivalent in drug substitution (∼ 6), cell binding (K(d) ∼ 1.2 nmol/L), cytotoxicity (IC(50) ∼ 2.2 nmol/L), and serum stability in vitro (t/(½) ∼ 20 hours). Exposure of cells to the ADC demonstrated signaling pathways leading to PARP cleavage, but differences versus free SN-38 in p53 and p21 upregulation were noted. Significant antitumor effects were produced by hRS7-SN-38 at nontoxic doses in mice bearing Calu-3 (P ≤ 0.05), Capan-1 (P < 0.018), BxPC-3 (P < 0.005), and COLO 205 tumors (P < 0.033) when compared to nontargeting control ADCs. Mice tolerated a dose of 2 × 12 mg/kg (SN-38 equivalents) with only short-lived elevations in ALT and AST liver enzyme levels. Cynomolgus monkeys infused with 2 × 0.96 mg/kg exhibited only transient decreases in blood counts, although, importantly, the values did not fall below normal ranges. CONCLUSIONS The anti-Trop-2 hRS7-CL2A-SN-38 ADC provides significant and specific antitumor effects against a range of human solid tumor types. It is well tolerated in monkeys, with tissue Trop-2 expression similar to humans, at clinically relevant doses, and warrants clinical investigation.
Collapse
Affiliation(s)
- Thomas M. Cardillo
- Immunomedics, Inc., Morris Plains, NJ 07950
- Contributed equally to this work
| | | | - Robert M. Sharkey
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Belleville, NJ 07109
| | | | - David M. Goldenberg
- Center for Molecular Medicine and Immunology, Garden State Cancer Center, Belleville, NJ 07109
| |
Collapse
|
19
|
Slagsvold JE, Pettersen CHH, Størvold GL, Follestad T, Krokan HE, Schønberg SA. DHA alters expression of target proteins of cancer therapy in chemotherapy resistant SW620 colon cancer cells. Nutr Cancer 2010; 62:611-21. [PMID: 20574922 DOI: 10.1080/01635580903532366] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Diets rich in n-3 polyunsaturated fatty acids (PUFAs) have been associated with a reduced risk of several types of cancer. Recent reports have suggested that these PUFAs enhance the cytotoxic effect of cancer chemoradiotherapy. The effect of docosahexaenoic acid (DHA) on key cell cycle regulators and target proteins of cancer therapy was investigated in the human malign colon cancer cell line SW620. Cell cycle check point proteins such as p21 and stratifin (14-3-3 sigma) increased at mRNA and protein level, whereas cell cycle progression proteins such as cell division cycle 25 homolog and cyclin-dependent kinase 1 decreased after DHA treatment. Protein levels of inhibitors of apoptosis family members associated with chemotherapy resistance and cancer malignancy, survivin and livin, decreased after the same treatment: likewise the expression of NF-kappaB. Levels of the proapoptotic proteins phosphorylated p38 MAPK and growth arrest-inducible and DNA damage-inducible gene 153/C/EBP-homologous protein (CHOP) increased. The results indicate that DHA treatment causes simultaneous cell cycle arrest in both the G1 and G2 phase. In conclusion, DHA affects several target proteins of chemotherapy in a favorable way. This may explain the observed enhanced chemosensitivity in cancer cells supplemented with n-3 PUFAs and encourage further studies investigating the role of n-3 PUFAs as adjuvant to chemotherapy and radiotherapy in vivo.
Collapse
Affiliation(s)
- Jens E Slagsvold
- Norwegian University of Science and Technology, Trondheim, Norway
| | | | | | | | | | | |
Collapse
|
20
|
Sensitivity and gene expression profile of fresh human acute myeloid leukemia cells exposed ex vivo to AS602868. Cancer Chemother Pharmacol 2010; 68:97-105. [PMID: 20844879 DOI: 10.1007/s00280-010-1458-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Accepted: 09/02/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE The need for new treatment options for acute myeloid leukemia (AML) is increasing. AS602868 is a novel investigational drug with reported activity on AML cells. METHODS We studied gene expression profiles in AML blasts exposed to AS602868 in order to better understand its mechanism of action. We analyzed the in vitro cytotoxicity of AS602868 alone or in combination with daunorubicin, etoposide or cytarabine. To document AS602868-induced IKK2 inhibition in fresh AML cells, a flow cytometry analysis of IκB was performed. Finally, the effect of AS602868 on gene expression in fresh AML cells was analyzed. RESULTS The results show that AML cells are globally as sensitive to AS602868 as they are to cytarabine, with large interindividual variations. Combinations with conventional antileukemic agents showed enhanced cytotoxic activity in subsets of patients. IKK2 appeared to be effectively inhibited by 100 μM AS602868 in fresh leukemic cells. Gene expression profiling and gene ontology analyses identified several groups of genes induced/inhibited by exposure to AS602868 and/or exhibiting a correlation with sensitivity to this agent in vitro. Of note, the expression of several genes related to immune function was found to be significantly altered after exposure to AS602868. CONCLUSION These data suggest that AS602868 is cytotoxic against fresh human AML blasts and provide insights regarding the mechanisms of cytotoxicity.
Collapse
|
21
|
Yang Z, Song L, Huang C. Gadd45 proteins as critical signal transducers linking NF-kappaB to MAPK cascades. Curr Cancer Drug Targets 2009; 9:915-30. [PMID: 20025601 PMCID: PMC3762688 DOI: 10.2174/156800909790192383] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The growth arrest and DNA damage-inducible 45 (Gadd45) proteins are a group of critical signal transducers that are involved in regulations of many cellular functions. Accumulated data indicate that all three Gadd45 proteins (i.e., Gadd45alpha, Gadd45beta, and Gadd45gamma) play essential roles in connecting an upstream sensor module, the transcription Nuclear Factor-kappaB (NF-kappaB), to a transcriptional regulating module, mitogen-activated protein kinase (MAPK). This NF-kappaB-Gadd45(s)-MAPK pathway responds to various kinds of extracellular stimuli and regulates such cell activities as growth arrest, differentiation, cell survival, and apoptosis. Defects in this pathway can also be related to oncogenesis. In the first part of this review, the functions of Gadd45 proteins, and briefly NF-kappaB and MAPK, are summarized. In the second part, the mechanisms by which Gadd45 proteins are regulated by NF-kappaB, and how they affect MAPK activation, are reviewed.
Collapse
Affiliation(s)
- Z. Yang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | - L. Song
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
- Department of Cellular Immunology, Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Beijing 100850, China
| | - C. Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| |
Collapse
|
22
|
Apoptosis and colorectal cancer: implications for therapy. Trends Mol Med 2009; 15:225-33. [PMID: 19362056 DOI: 10.1016/j.molmed.2009.03.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 03/09/2009] [Accepted: 03/09/2009] [Indexed: 12/26/2022]
Abstract
Colorectal cancer (CRC) is characterized by the partial suppression of apoptosis, which in turn gives tumours a selective advantage for survival and can cause current chemotherapy approaches to be ineffective. Recent progress in understanding the mechanisms of apoptosis in colorectal carcinogenesis has provided potential new targets for therapy. Here, we review recent studies of the regulation of apoptosis and its role in CRC initiation and progression, and we discuss the relationship between chemoresistance and the suppression of apoptosis. Recent progress in targeting apoptotic pathways and their regulators provide strategies for the exploration of novel therapies for CRC.
Collapse
|
23
|
Mei Q, Cao Z, Chen Y. Gentamycin combined with sodium bicarbonate for prevention of irinotecan-induced diarrhea: An analysis of 52 cases. Shijie Huaren Xiaohua Zazhi 2008; 16:3354-3358. [DOI: 10.11569/wcjd.v16.i29.3354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the therapeutic effects of gentamycin combined with sodium bicarbonate for irinotecan-induced diarrhea.
METHODS: A total of 98 patients with stage Ⅳ cancers were allotted into prevention group (n = 52 patients) and control group (n = 46 patients) from July 2006 to May 2008 in our hospital. All patients received the chemotherapy including irinotecan. The prevention group received gentamycin combined with sodium bicarbonate before the use of irinotecan for 4 d, while the control group received routine treatment. Tropisetron was conventionally used to prevent the vomiting in both groups. All adverse effects were evaluated using NCI-CTC standard.
RESULTS: The use of gentamycin combined with sodium bicarbonate resulted in significantly higher stool pH value (t = 15.295, P < 0.05), significantly reduced the incidence of irinotecan-induced diarrhea (13.70% vs 34.83%, P < 0.001) and medications (Loperamide: χ2 = 23.844, P < 0.001; Smecta: χ2 = 25.054, P < 0.001; Somafostatin: χ2 = 8.561, P = 0.003; Fluid replacement: χ2 = 8.007; P = 0.005). At the same time, bone marrow suppression, nausea and vomiting and liver function were also improved in the prevention group (χ2 = 80.335, 60.339, 5.155, P <0.01 or 0.05). However, there were no significant differences in appetite, constipation, renal function between the two groups.
CONCLUSION: Gentamycin combined with sodium bicarbonate proves to be useful for preventing the diarrhea induced by irinotecan and is recommended for wide use.
Collapse
|