1
|
Lin Z, Hua G, Hu X. Lipid metabolism associated crosstalk: the bidirectional interaction between cancer cells and immune/stromal cells within the tumor microenvironment for prognostic insight. Cancer Cell Int 2024; 24:295. [PMID: 39174964 PMCID: PMC11342506 DOI: 10.1186/s12935-024-03481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024] Open
Abstract
Cancer is closely related to lipid metabolism, with the tumor microenvironment (TME) containing numerous lipid metabolic interactions. Cancer cells can bidirectionally interact with immune and stromal cells, the major components of the TME. This interaction is primarily mediated by fatty acids (FAs), cholesterol, and phospholipids. These interactions can lead to various physiological changes, including immune suppression, cancer cell proliferation, dissemination, and anti-apoptotic effects on cancer cells. The physiological modulation resulting from this lipid metabolism-associated crosstalk between cancer cells and immune/stromal cells provides valuable insights into cancer prognosis. A comprehensive literature review was conducted to examine the function of the bidirectional lipid metabolism interactions between cancer cells and immune/stromal cells within the TME, particularly how these interactions influence cancer prognosis. A novel autophagy-extracellular vesicle (EV) pathway has been proposed as a mediator of lipid metabolism interactions between cancer cells and immune cells/stromal cells, impacting cancer prognosis. As a result, different forms of lipid metabolism interactions have been described as being linked to cancer prognosis, including those mediated by the autophagy-EV pathway. In conclusion, understanding the bidirectional lipid metabolism interactions between cancer cells and stromal/immune cells in the TME can help develop more advanced prognostic approaches for cancer patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Guanxiang Hua
- Queen Mary College, Nanchang University, Nanchang, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, UK
| | - Xiaojuan Hu
- Queen Mary College, Nanchang University, Nanchang, China.
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
2
|
Owen JS, Clayton A, Pearson HB. Cancer-Associated Fibroblast Heterogeneity, Activation and Function: Implications for Prostate Cancer. Biomolecules 2022; 13:67. [PMID: 36671452 PMCID: PMC9856041 DOI: 10.3390/biom13010067] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The continuous remodeling of the tumor microenvironment (TME) during prostate tumorigenesis is emerging as a critical event that facilitates cancer growth, progression and drug-resistance. Recent advances have identified extensive communication networks that enable tumor-stroma cross-talk, and emphasized the functional importance of diverse, heterogeneous stromal fibroblast populations during malignant growth. Cancer-associated fibroblasts (CAFs) are a vital component of the TME, which mediate key oncogenic events including angiogenesis, immunosuppression, metastatic progression and therapeutic resistance, thus presenting an attractive therapeutic target. Nevertheless, how fibroblast heterogeneity, recruitment, cell-of-origin and differential functions contribute to prostate cancer remains to be fully delineated. Developing our molecular understanding of these processes is fundamental to developing new therapies and biomarkers that can ultimately improve clinical outcomes. In this review, we explore the current challenges surrounding fibroblast identification, discuss new mechanistic insights into fibroblast functions during normal prostate tissue homeostasis and tumorigenesis, and illustrate the diverse nature of fibroblast recruitment and CAF generation. We also highlight the promise of CAF-targeted therapies for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jasmine S. Owen
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Aled Clayton
- Tissue Microenvironment Group, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
3
|
Liu ZY, Zhang DY, Lin XH, Sun JL, Abuduwaili W, Zhang GC, Xu RC, Wang F, Yu XN, Shi X, Deng B, Dong L, Weng SQ, Zhu JM, Shen XZ, Liu TT. Nalidixic acid potentiates the antitumor activity in sorafenib-resistant hepatocellular carcinoma via the tumor immune microenvironment analysis. Front Pharmacol 2022; 13:952482. [PMID: 36071851 PMCID: PMC9441713 DOI: 10.3389/fphar.2022.952482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/21/2022] [Indexed: 12/24/2022] Open
Abstract
Sorafenib resistance is often developed and impedes the benefits of clinical therapy in hepatocellular carcinoma (HCC) patients. However, the relationship between sorafenib resistance and tumor immune environment and adjuvant drugs for sorafenib-resistant HCC are not systemically identified. This study first analyzed the expression profiles of sorafenib-resistant HCC cells to explore immune cell infiltration levels and differentially expressed immune-related genes (DEIRGs). The prognostic value of DEIRGs was analyzed using Cox regression and Kaplan-Meier analysis based on The Cancer Genome Atlas. The primary immune cells infiltrated in sorafenib-resistant HCC mice were explored using flow cytometry (FCM). Finally, small-molecule drugs for sorafenib-resistant HCC treatment were screened and validated by experiments. The CIBERSORT algorithm and mice model showed that macrophages and neutrophils are highly infiltrated, while CD8+ T cells are downregulated in sorafenib-resistant HCC. Totally, 34 DEIRGs were obtained from sorafenib-resistant and control groups, which were highly enriched in immune-associated biological processes and pathways. NR6A1, CXCL5, C3, and TGFB1 were further identified as prognostic markers for HCC patients. Finally, nalidixic acid was identified as a promising antagonist for sorafenib-resistant HCC treatment. Collectively, our study reveals the tumor immune microenvironment changes and explores a promising adjuvant drug to overcome sorafenib resistance in HCC.
Collapse
Affiliation(s)
- Zhi-Yong Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Dan-Ying Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Xia-Hui Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Jia-Lei Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Weinire Abuduwaili
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Guang-Cong Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Ru-Chen Xu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Fu Wang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Xiang-Nan Yu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Xuan Shi
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Bin Deng
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Ling Dong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Shu-Qiang Weng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Ji-Min Zhu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| | - Xi-Zhong Shen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
- Key Laboratory of Medical Molecular Virology, Shanghai Medical College of Fudan University, Shanghai, China
| | - Tao-Tao Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital of Fudan University, Shanghai, China
- Shanghai Institute of Liver Disease, Shanghai, China
| |
Collapse
|
4
|
Tilsed CM, Fisher SA, Nowak AK, Lake RA, Lesterhuis WJ. Cancer chemotherapy: insights into cellular and tumor microenvironmental mechanisms of action. Front Oncol 2022; 12:960317. [PMID: 35965519 PMCID: PMC9372369 DOI: 10.3389/fonc.2022.960317] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy has historically been the mainstay of cancer treatment, but our understanding of what drives a successful therapeutic response remains limited. The diverse response of cancer patients to chemotherapy has been attributed principally to differences in the proliferation rate of the tumor cells, but there is actually very little experimental data supporting this hypothesis. Instead, other mechanisms at the cellular level and the composition of the tumor microenvironment appear to drive chemotherapy sensitivity. In particular, the immune system is a critical determinant of chemotherapy response with the depletion or knock-out of key immune cell populations or immunological mediators completely abrogating the benefits of chemotherapy in pre-clinical models. In this perspective, we review the literature regarding the known mechanisms of action of cytotoxic chemotherapy agents and the determinants of response to chemotherapy from the level of individual cells to the composition of the tumor microenvironment. We then summarize current work toward the development of dynamic biomarkers for response and propose a model for a chemotherapy sensitive tumor microenvironment.
Collapse
Affiliation(s)
- Caitlin M. Tilsed
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Scott A. Fisher
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - Anna K. Nowak
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- Medical School, University of Western Australia, Crawley, WA, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Richard A. Lake
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
| | - W. Joost Lesterhuis
- National Centre for Asbestos Related Diseases, Institute for Respiratory Health, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Crawley, WA, Australia
- Telethon Kids Institute, University of Western Australia, West Perth, WA, Australia
- *Correspondence: W. Joost Lesterhuis,
| |
Collapse
|
5
|
Rijal G. Understanding the Role of Fibroblasts following a 3D Tumoroid Implantation for Breast Tumor Formation. Bioengineering (Basel) 2021; 8:bioengineering8110163. [PMID: 34821729 PMCID: PMC8615023 DOI: 10.3390/bioengineering8110163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
An understanding of the participation and modulation of fibroblasts during tumor formation and growth is still unclear. Among many speculates, one might be the technical challenge to reveal the versatile function of fibroblasts in tissue complexity, and another is the dynamics in tissue physiology and cell activity. The histology of most solid tumors shows a predominant presence of fibroblasts, suggesting that tumor cells recruit fibroblasts for breast tumor growth. In this review paper, therefore, the migration, activation, differentiation, secretion, and signaling systems that are associated with fibroblasts and cancer-associated fibroblasts (CAFs) after implantation of a breast tumoroid, i.e., a lab-generated tumor tissue into an animal, are discussed.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Medical Laboratory Sciences and Public Health, Tarleton State University, a Member of Texas A & M University System, Fort Worth, TX 76104, USA
| |
Collapse
|
6
|
León-González AJ, Sáez-Martínez P, Jiménez-Vacas JM, Herrero-Aguayo V, Montero-Hidalgo AJ, Gómez-Gómez E, Madrona A, Castaño JP, Espartero JL, Gahete MD, Luque RM. Comparative Cytotoxic Activity of Hydroxytyrosol and Its Semisynthetic Lipophilic Derivatives in Prostate Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10091348. [PMID: 34572980 PMCID: PMC8464900 DOI: 10.3390/antiox10091348] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/21/2021] [Indexed: 12/30/2022] Open
Abstract
A high adherence to a Mediterranean diet has been related to numerous beneficial effects in human health, including a lower incidence and mortality of prostate cancer (PCa). Olive oil is an important source of phenolic bioactive compounds, mainly hydroxytyrosol (HT), of this diet. Because of the growing interest of this compound and its derivatives as a cancer chemopreventive agent, we aimed to compare the in vitro effect of HT isolated from olive mill wastewaters and five semisynthetic alkyl ether, ester, and nitro-derivatives against prostate cancer (PCa) cell lines. The effect in cell proliferation was determined in RWPE-1, LNCaP, 22Rv1, and PC-3 cells by resazurin assay, the effect in cell migration by wound healing assay, and tumorsphere and colony formation were evaluated. The changes in key signaling pathways involved in carcinogenesis were assessed by using a phosphorylation pathway profiling array and by Western blotting. Antiproliferative effects of HT and two lipophilic derivatives [hydroxytyrosyl acetate (HT-Ac)/ethyl hydroxytyrosyl ether (HT-Et)] were significantly higher in cancerous PC-3 and 22Rv1 cells than in non-malignant RWPE-1 cells. HT/HT-Ac/HT-Et significantly reduced migration capacity in RWPE-1 and PC-3 and prostatosphere size and colony formation in 22Rv1, whereas only HT-Ac and HT-Et reduced these functional parameters in PC-3. The cytotoxic effect in 22Rv1 cells was correlated with modifications in the phosphorylation pattern of key proteins, including ERK1/2 and AKT. Consistently, HT-Ac and HT-Et decreased p-AKT levels in PC-3. In sum, our results suggest that HT and its lipophilic derivatives could be considered as potential therapeutic tools in PCa.
Collapse
Affiliation(s)
- Antonio J. León-González
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain
- Correspondence: (A.J.L.-G.); (R.M.L.); Tel.: +34-957213740 (R.M.L.)
| | - Prudencio Sáez-Martínez
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Juan M. Jiménez-Vacas
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Vicente Herrero-Aguayo
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Antonio J. Montero-Hidalgo
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Enrique Gómez-Gómez
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- Urology Service, HURS/IMIBIC, 14004 Cordoba, Spain
| | - Andrés Madrona
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (A.M.); (J.L.E.)
| | - Justo P. Castaño
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - José L. Espartero
- Department of Organic and Pharmaceutical Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (A.M.); (J.L.E.)
| | - Manuel D. Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
| | - Raúl M. Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), 14004 Cordoba, Spain; (P.S.-M.); (J.M.J.-V.); (V.H.-A.); (A.J.M.-H.); (E.G.-G.); (J.P.C.); (M.D.G.)
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14014 Cordoba, Spain
- Reina Sofia University Hospital (HURS), 14004 Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), 14004 Cordoba, Spain
- Correspondence: (A.J.L.-G.); (R.M.L.); Tel.: +34-957213740 (R.M.L.)
| |
Collapse
|
7
|
Dahiya M, Dureja H. Sorafenib for hepatocellular carcinoma: potential molecular targets and resistance mechanisms. J Chemother 2021; 34:286-301. [PMID: 34291704 DOI: 10.1080/1120009x.2021.1955202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most widespread typical therapy-resistant, unresectable type of malignant solid tumour with a high death rate constituting huge medical concern. Sorafenib is a small molecule oral multi-target kinase potent inhibitor that acts by suppressing/blocking the multiplication of the tumour cells, angiogenesis, and encouraging apoptosis of the tumour cells. Though, the precise mechanism of tumour cell death induction by sorafenib is yet under exploration. Furthermore, genetic heterogeneity plays a critical role in developing sorafenib resistance, which leads the way to identify the need for predictive biomarkers responsible for drug resistance. Therefore, it is essential to find out the fundamental resistance mechanisms to expand therapeutic plans. The authors summarize the molecular concepts of resistance, progression, potential molecular targets, HCC management therapies, and discussion on the advancements expected in the coming future, inclusive of biomarker-driven treatment strategies, which may provide the prospects to design innovative therapeutically targeted strategies for the HCC treatment and the clinical implementation of emerging targeted agents.
Collapse
Affiliation(s)
- Mandeep Dahiya
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
8
|
Wu F, Yang J, Liu J, Wang Y, Mu J, Zeng Q, Deng S, Zhou H. Signaling pathways in cancer-associated fibroblasts and targeted therapy for cancer. Signal Transduct Target Ther 2021; 6:218. [PMID: 34108441 PMCID: PMC8190181 DOI: 10.1038/s41392-021-00641-0] [Citation(s) in RCA: 389] [Impact Index Per Article: 97.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/20/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
To flourish, cancers greatly depend on their surrounding tumor microenvironment (TME), and cancer-associated fibroblasts (CAFs) in TME are critical for cancer occurrence and progression because of their versatile roles in extracellular matrix remodeling, maintenance of stemness, blood vessel formation, modulation of tumor metabolism, immune response, and promotion of cancer cell proliferation, migration, invasion, and therapeutic resistance. CAFs are highly heterogeneous stromal cells and their crosstalk with cancer cells is mediated by a complex and intricate signaling network consisting of transforming growth factor-beta, phosphoinositide 3-kinase/AKT/mammalian target of rapamycin, mitogen-activated protein kinase, Wnt, Janus kinase/signal transducers and activators of transcription, epidermal growth factor receptor, Hippo, and nuclear factor kappa-light-chain-enhancer of activated B cells, etc., signaling pathways. These signals in CAFs exhibit their own special characteristics during the cancer progression and have the potential to be targeted for anticancer therapy. Therefore, a comprehensive understanding of these signaling cascades in interactions between cancer cells and CAFs is necessary to fully realize the pivotal roles of CAFs in cancers. Herein, in this review, we will summarize the enormous amounts of findings on the signals mediating crosstalk of CAFs with cancer cells and its related targets or trials. Further, we hypothesize three potential targeting strategies, including, namely, epithelial-mesenchymal common targets, sequential target perturbation, and crosstalk-directed signaling targets, paving the way for CAF-directed or host cell-directed antitumor therapy.
Collapse
Affiliation(s)
- Fanglong Wu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Junjiang Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Ye Wang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jingtian Mu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Qingxiang Zeng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Shuzhi Deng
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
9
|
Turnham DJ, Bullock N, Dass MS, Staffurth JN, Pearson HB. The PTEN Conundrum: How to Target PTEN-Deficient Prostate Cancer. Cells 2020; 9:E2342. [PMID: 33105713 PMCID: PMC7690430 DOI: 10.3390/cells9112342] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/17/2022] Open
Abstract
Loss of the tumor suppressor phosphatase and tensin homologue deleted on chromosome 10 (PTEN), which negatively regulates the PI3K-AKT-mTOR pathway, is strongly linked to advanced prostate cancer progression and poor clinical outcome. Accordingly, several therapeutic approaches are currently being explored to combat PTEN-deficient tumors. These include classical inhibition of the PI3K-AKT-mTOR signaling network, as well as new approaches that restore PTEN function, or target PTEN regulation of chromosome stability, DNA damage repair and the tumor microenvironment. While targeting PTEN-deficient prostate cancer remains a clinical challenge, new advances in the field of precision medicine indicate that PTEN loss provides a valuable biomarker to stratify prostate cancer patients for treatments, which may improve overall outcome. Here, we discuss the clinical implications of PTEN loss in the management of prostate cancer and review recent therapeutic advances in targeting PTEN-deficient prostate cancer. Deepening our understanding of how PTEN loss contributes to prostate cancer growth and therapeutic resistance will inform the design of future clinical studies and precision-medicine strategies that will ultimately improve patient care.
Collapse
Affiliation(s)
- Daniel J. Turnham
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - Nicholas Bullock
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Manisha S. Dass
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| | - John N. Staffurth
- Division of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK;
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK; (D.J.T.); (N.B.); (M.S.D.)
| |
Collapse
|
10
|
Binary blended co-delivery nanoparticles with the characteristics of precise pH-responsive acting on tumor microenvironment. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 117:111370. [PMID: 32919698 DOI: 10.1016/j.msec.2020.111370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/19/2020] [Accepted: 07/30/2020] [Indexed: 01/12/2023]
Abstract
Although combined chemotherapy had achieved the ideal efficacy in clinical anti-cancer therapeutic, the issues that need to be addressed are non-targeting and toxic-side effects of small molecule chemical drug (SMCD). In this study, we designed and prepared a novel binary blended co-delivered nanoparticles (BBCD NPs) with pH-responsive feature on tumor microenvironment. The BBCD NPs consists of two kind of drug-loaded NPs, in one of which carboxymethyl chitosan (CMC) and Poly (lactic-co-glycolic acid) (PLGA) were chosen as delivery carrier to load anti-cancer drug vincristine (VCR), named CMC-PLGA-VCR NPs (or CPNPVCR); and in the other of which methoxy poly(ethylene glycol)-poly(β-amino ester) (mPEG-PAE) were chosen as delivery carrier to load anti-fibrotic drug pirfenidone (PFD), named mPEG-PAE-PFD NPs (or PPNPPFD). Then, the two types of NPs (CPNPVCR and PPNPPFD) were physically mixed in mass ratios to form BBCD NPs, which was named CPNPVCR&PPNPPFD. CPNPVCR&PPNPPFD had good encapsulation efficiency and loading capacity, and the particle size distribution was uniform. In cytotoxicity experiments and non-contact co-culture studies in vitro, the model drugs loaded in CPNPVCR&PPNPPFD could respectively target cancer cell and cancer associated fibroblast (CAF) owing to the precise pH-sensitive drug release in the pharmacological targets and show stronger synergism than that of the combined treatment of two free drugs. As a modularity and assemble ability feature in design, BBCD NPs would have the advantages on the terms of concise on preparation process, controllable on quality standard, stable in natural environment storage. The research results can provide scientific evidence for the further development of a novel drug co-delivery system with multi-type cell targets.
Collapse
|
11
|
Cheteh EH, Sarne V, Ceder S, Bianchi J, Augsten M, Rundqvist H, Egevad L, Östman A, Wiman KG. Interleukin-6 derived from cancer-associated fibroblasts attenuates the p53 response to doxorubicin in prostate cancer cells. Cell Death Discov 2020; 6:42. [PMID: 32528731 PMCID: PMC7265343 DOI: 10.1038/s41420-020-0272-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) promote tumor growth and progression, and increase drug resistance through several mechanisms. We have investigated the effect of CAFs on the p53 response to doxorubicin in prostate cancer cells. We show that CAFs produce interleukin-6 (IL-6), and that IL-6 attenuates p53 induction and upregulation of the pro-apoptotic p53 target Bax upon treatment with doxorubicin. This is associated with increased levels of MDM2 mRNA, Mdm2 protein bound to p53, and ubiquitinated p53. IL-6 also inhibited doxorubicin-induced cell death. Inhibition of JAK or STAT3 alleviated this effect, indicating that IL-6 attenuates p53 via the JAK/STAT signaling pathway. These results suggest that CAF-derived IL-6 plays an important role in protecting cancer cells from chemotherapy and that inhibition of IL-6 could have significant therapeutic value.
Collapse
Affiliation(s)
| | - Victoria Sarne
- Department of Pathobiochemistry and Genetics, Medical University of Vienna, Vienna, Austria
| | - Sophia Ceder
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Julie Bianchi
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Martin Augsten
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Klas G. Wiman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Yang ZY, Wang Y, Liu Q, Wu M. microRNA cluster MC-let-7a-1~let-7d promotes autophagy and apoptosis of glioma cells by down-regulating STAT3. CNS Neurosci Ther 2019; 26:319-331. [PMID: 31868319 PMCID: PMC7052808 DOI: 10.1111/cns.13273] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 10/30/2019] [Accepted: 11/13/2019] [Indexed: 12/12/2022] Open
Abstract
Background Accumulating evidence has highlighted the correlation between microRNAs (miRNAs) and the progression of glioma. However, the role of miR cluster MC‐let‐7a‐1 ~ let‐7d in glioma remains elusive. Thus, the current study aimed to investigate the effect of miR cluster MC‐let‐7a‐1 ~ let‐7d on glioma progression. Methods and Results Microarray data analysis provided data indicating the involvement of miR cluster MC‐let‐7a‐1 ~ let‐7d in glioma via STAT3. The expression of let‐7a‐1, let‐7d, let‐7f‐1, and miR cluster MC‐let‐7a‐1 ~ let‐7d was diminished in the glioma tissues and the cell lines. Additionally, our results revealed that STAT3 was a target gene of let‐7d, let‐7a‐1, and let‐7f‐1, which was further verified by the dual‐luciferase reporter gene assay. Moreover, STAT3 expression was negatively mediated by let‐7a‐1, let‐7d, and let‐7f‐1. Up‐regulated miR cluster MC‐let‐7a‐1 ~ let‐7d or silenced STAT3 suppressed cell proliferation but accelerated cell apoptosis and autophagy. Moreover, restrained tumor growth was identified in the nude mice treated with miR cluster MC‐let‐7a‐1 ~ let‐7d mimics or STAT3 siRNA. Conclusion Taken together, the miR cluster MC‐let‐7a‐1 ~ let‐7d promotes glioma cell autophagy and apoptosis by repressing STAT3. The current study highlights the potential of the miR cluster MC‐let‐7a‐1 ~ let‐7d as biomarkers and promising treatment strategies for glioma.
Collapse
Affiliation(s)
- Zhuan-Yi Yang
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, China
| | - Ying Wang
- Department of Pathology, Xiangya Medical School of Central South University & Xiangya Hospital Central South University, Changsha, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, China
| | - Ming Wu
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
13
|
Lin HP, Ho HM, Chang CW, Yeh SD, Su YW, Tan TH, Lin WJ. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis. FASEB J 2019; 33:14653-14667. [PMID: 31693867 DOI: 10.1096/fj.201802558rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dual-specificity phosphatases (DUSPs) regulate the activity of various downstream kinases through serine or threonine or tyrosine dephosphorylation. Loss of function and aberrant expression of DUSPs has been implicated in cancer progression and poor survival, yet the function of DUSP22 in prostate cancer (PCa) cells is not clear. Gene Expression Omnibus and cBioPortal microarray database analyses showed that DUSP22 expression was lower in PCa tissues than normal prostate tissues, and altered DUSP22 expression was associated with shorter progression-free and disease-free survival of patients with PCa. Exogenous DUSP22 expression in LNCaP, PC3, and C4-2B PCa cells inhibited cellular proliferation and colony formation, supporting a growth inhibitory role for DUSP22 in PCa cells. DUSP22 expression significantly attenuated epidermal growth factor (EGF) receptor (EGFR) and its downstream ERK1/2 signaling by dephosphorylation. However, DUSP22 failed to suppress the growth of CWR22Rv1 and DU145 cells with elevated phosphorylated (p-)ERK1/2 levels. A serine-to-alanine mutation at position 58, a potential ERK1/2-targeted phosphorylation site in DUSP22, was sufficient to suppress growth of CWR22Rv1 cells with elevated p-ERK1/2 levels, suggesting a mutually antagonistic relationship between DUSP22 and ERK1/2 dependent on phosphorylation status. We showed that DUSP22 can suppress prostate-specific antigen gene expression through phosphatase-dependent pathways, suggesting that DUSP22 is an important regulator of the androgen receptor (AR) in PCa cells. Mechanistically, DUSP22 can interact with AR as a regulatory partner and interfere with EGF-induced AR phosphorylation at Tyr534, suggesting that DUSP22 serves as a crucial suppressor of both EGFR and AR-dependent signaling in PCa cells via dephosphorylation. Our findings indicate that loss of function of DUSP22 in PCa cells leads to aberrant activation of both EGFR-ERKs and AR signaling and ultimately progression of PCa, supporting the potential for novel therapeutic design of harnessing DUSP22 in the treatment of PCa.-Lin, H.-P., Ho, H.-M., Chang, C.-W., Yeh, S.-D., Su, Y.-W., Tan, T.-H., Lin, W.-J. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis.
Collapse
Affiliation(s)
- Hsiu-Ping Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Hui-Min Ho
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Cheng-Wei Chang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Shauh-Der Yeh
- Department of Urology, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Wen-Jye Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| |
Collapse
|
14
|
Qu Y, Dou B, Tan H, Feng Y, Wang N, Wang D. Tumor microenvironment-driven non-cell-autonomous resistance to antineoplastic treatment. Mol Cancer 2019; 18:69. [PMID: 30927928 PMCID: PMC6441162 DOI: 10.1186/s12943-019-0992-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/28/2019] [Indexed: 12/24/2022] Open
Abstract
Drug resistance is of great concern in cancer treatment because most effective drugs are limited by the development of resistance following some periods of therapeutic administration. The tumor microenvironment (TME), which includes various types of cells and extracellular components, mediates tumor progression and affects treatment efficacy. TME-mediated drug resistance is associated with tumor cells and their pericellular matrix. Noninherent-adaptive drug resistance refers to a non-cell-autonomous mechanism in which the resistance lies in the treatment process rather than genetic or epigenetic changes, and this mechanism is closely related to the TME. A new concept is therefore proposed in which tumor cell resistance to targeted therapy may be due to non-cell-autonomous mechanisms. However, knowledge of non-cell-autonomous mechanisms of resistance to different treatments is not comprehensive. In this review, we outlined TME factors and molecular events involved in the regulation of non-cell-autonomous resistance of cancer, summarized how the TME contributes to non-cell-autonomous drug resistance in different types of antineoplastic treatment, and discussed the novel strategies to investigate and overcome the non-cell-autonomous mechanism of cancer non-cell-autonomous resistance.
Collapse
Affiliation(s)
- Yidi Qu
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Bo Dou
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Horyue Tan
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, 130012, China.
- School of Chinese Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
15
|
Levesque C, Nelson PS. Cellular Constituents of the Prostate Stroma: Key Contributors to Prostate Cancer Progression and Therapy Resistance. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a030510. [PMID: 28490538 DOI: 10.1101/cshperspect.a030510] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Reciprocal signaling between prostate stroma and its epithelium are fundamental to organ development and homeostasis. Similarly, interactions between tumor cells and stromal constituents are central to key aspects of carcinogenesis and malignancy growth involving tumor cell invasion, dissemination, and growth in distant sites. The prostate stroma is complex with several distinct resident cell types, infiltrating nonresident cell types and an amalgam of structural matrix factors, matricellular proteins, metabolites, growth factors, and cytokines. Of importance, the stroma is dynamic with changes in composition as a cause or consequence of intrinsic and extrinsic factors. In the context of epithelial neoplasia, the prostate stroma undergoes phenotypic changes with a loss of well-differentiated smooth muscle cell population and the expansion of cancer-associated fibroblast populations. This reactive stroma further coevolves with tumor progression. Recent studies show the role of tumor microenvironment components in therapy resistance and highlight the importance of a thorough knowledge of cross talk between tumor cells and microenvironment niches to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Christine Levesque
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
| |
Collapse
|
16
|
Bad phosphorylation as a target of inhibition in oncology. Cancer Lett 2017; 415:177-186. [PMID: 29175460 DOI: 10.1016/j.canlet.2017.11.017] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/13/2017] [Accepted: 11/17/2017] [Indexed: 12/19/2022]
Abstract
Bcl-2 agonist of cell death (BAD) is a BH3-only member of the Bcl-2 family which possesses important regulatory function in apoptosis. BAD has also been shown to possess many non-apoptotic functions closely linked to cancer including regulation of glycolysis, autophagy, cell cycle progression and immune system development. Interestingly, BAD can be either pro-apoptotic or pro-survival depending on the phosphorylation state of three specific serine residues (human S75, S99 and S118). Expression of BAD and BAD phosphorylation patterns have been shown to influence tumor initiation and progression and play a predictive role in disease prognosis, drug response and chemosensitivity in various cancers. This review aims to summarize the current evidence on the functional role of BAD phosphorylation in human cancer and evaluate the potential utility of modulating BAD phosphorylation in cancer.
Collapse
|
17
|
Lin CL, Chen CM, Lin CL, Cheng CW, Lee CH, Hsieh YH. Norcantharidin induces mitochondrial-dependent apoptosis through Mcl-1 inhibition in human prostate cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1867-1876. [PMID: 28760656 DOI: 10.1016/j.bbamcr.2017.07.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/20/2017] [Accepted: 07/27/2017] [Indexed: 12/12/2022]
Abstract
Norcantharidin (NCTD) is the demethylated form of cantharidin that exhibits anticancer potential in many cancer cell types. Recent reports suggest that NCTD targeting ROS/AMPK and DNA replication signaling pathway could be an effective strategy for the treatment of PCa cells. However, supportive evidence is limited to the effect of NCTD that induction of apoptosis through suppression of the Mcl-1. Here, we show that NCTD induced PCa cell apoptosis and triggered caspase activation, which was associated with mitochondria dysfunction. Mechanistic investigations suggested that NCTD modulated the Akt signaling via increased nuclear translocation and interaction with the myeloid cell leukemia-1 (Mcl-1) promoter by FOXO4, resulting in an apoptotic effect. Moreover, miR-320d, which targets Mcl-1, was significantly upregulated after NCTD treatment. Overexpression of miR-320d by NCTD induced mitochondria dysfunction and apoptosis, which was notably attenuated with a miR-320d inhibitor. In vivo xenograft analysis revealed that NCTD significantly reduced tumor growth in mice with PC3 tumor xenografts. Taken together, our results provide new insights into the critical role of NCTD in suppressing Mcl-1 via epigenetic upregulation of miR-320d, resulting in PCa cell apoptosis.
Collapse
Affiliation(s)
- Chu-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan; School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Hsing Lee
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Division of Pediatric Surgery, Department of Surgery, Children's Hospital of China Medical University, Taichung. Taiwan.
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung, Taiwan; Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Clinical laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
18
|
Cheteh EH, Augsten M, Rundqvist H, Bianchi J, Sarne V, Egevad L, Bykov VJ, Östman A, Wiman KG. Human cancer-associated fibroblasts enhance glutathione levels and antagonize drug-induced prostate cancer cell death. Cell Death Dis 2017; 8:e2848. [PMID: 28569790 PMCID: PMC5520886 DOI: 10.1038/cddis.2017.225] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 03/30/2017] [Accepted: 04/20/2017] [Indexed: 11/15/2022]
Abstract
Drug resistance is a major problem in cancer therapy. A growing body of evidence demonstrates that the tumor microenvironment, including cancer-associated fibroblasts (CAFs), can modulate drug sensitivity in tumor cells. We examined the effect of primary human CAFs on p53 induction and cell viability in prostate cancer cells on treatment with chemotherapeutic drugs. Co-culture with prostate CAFs or CAF-conditioned medium attenuated DNA damage and the p53 response to chemotherapeutic drugs and enhanced prostate cancer cell survival. CAF-conditioned medium inhibited the accumulation of doxorubicin, but not taxol, in prostate cancer cells in a manner that was associated with increased cancer cell glutathione levels. A low molecular weight fraction (<3 kDa) of CAF-conditioned medium had the same effect. CAF-conditioned medium also inhibited induction of reactive oxygen species (ROS) in both doxorubicin- and taxol-treated cancer cells. Our findings suggest that CAFs can enhance drug resistance in cancer cells by inhibiting drug accumulation and counteracting drug-induced oxidative stress. This protective mechanism may represent a novel therapeutic target in cancer.
Collapse
Affiliation(s)
- Emarndeena H Cheteh
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Martin Augsten
- Division for Vascular Oncology and Metastasis, German Cancer Research Center, Heidelberg, Germany
| | - Helene Rundqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Julie Bianchi
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Victoria Sarne
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Lars Egevad
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Jn Bykov
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Klas G Wiman
- Department of Oncology-Pathology, Cancer Center Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
19
|
Xu Z, Yang F, Wei D, Liu B, Chen C, Bao Y, Wu Z, Wu D, Tan H, Li J, Wang J, Liu J, Sun S, Qu L, Wang L. Long noncoding RNA-SRLR elicits intrinsic sorafenib resistance via evoking IL-6/STAT3 axis in renal cell carcinoma. Oncogene 2016; 36:1965-1977. [DOI: 10.1038/onc.2016.356] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 07/25/2016] [Accepted: 08/15/2016] [Indexed: 12/28/2022]
|
20
|
Kharaziha P, Chioureas D, Baltatzis G, Fonseca P, Rodriguez P, Gogvadze V, Lennartsson L, Björklund AC, Zhivotovsky B, Grandér D, Egevad L, Nilsson S, Panaretakis T. Sorafenib-induced defective autophagy promotes cell death by necroptosis. Oncotarget 2016; 6:37066-82. [PMID: 26416459 PMCID: PMC4741916 DOI: 10.18632/oncotarget.5797] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Accepted: 08/29/2015] [Indexed: 01/06/2023] Open
Abstract
Autophagy is one of the main cytoprotective mechanisms that cancer cells deploy to withstand the cytotoxic stress and survive the lethal damage induced by anti-cancer drugs. However, under specific conditions, autophagy may, directly or indirectly, induce cell death. In our study, treatment of the Atg5-deficient DU145 prostate cancer cells, with the multi-tyrosine kinase inhibitor, sorafenib, induces mitochondrial damage, autophagy and cell death. Molecular inhibition of autophagy by silencing ULK1 and Beclin1 rescues DU145 cells from cell death indicating that, in this setting, autophagy promotes cell death. Re-expression of Atg5 restores the lipidation of LC3 and rescues DU145 and MEF atg5−/− cells from sorafenib-induced cell death. Despite the lack of Atg5 expression and LC3 lipidation, DU145 cells form autophagosomes as demonstrated by transmission and immuno-electron microscopy, and the formation of LC3 positive foci. However, the lack of cellular content in the autophagosomes, the accumulation of long-lived proteins, the presence of GFP-RFP-LC3 positive foci and the accumulated p62 protein levels indicate that these autophagosomes may not be fully functional. DU145 cells treated with sorafenib undergo a caspase-independent cell death that is inhibited by the RIPK1 inhibitor, necrostatin-1. Furthermore, treatment with sorafenib induces the interaction of RIPK1 with p62, as demonstrated by immunoprecipitation and a proximity ligation assay. Silencing of p62 decreases the RIPK1 protein levels and renders necrostatin-1 ineffective in blocking sorafenib-induced cell death. In summary, the formation of Atg5-deficient autophagosomes in response to sorafenib promotes the interaction of p62 with RIPK leading to cell death by necroptosis.
Collapse
Affiliation(s)
- Pedram Kharaziha
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Dimitris Chioureas
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - George Baltatzis
- Department of Medicine, School of Health Sciences, University of Athens, Athens, Greece
| | - Pedro Fonseca
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Patricia Rodriguez
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Gogvadze
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Lena Lennartsson
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Ann-Charlotte Björklund
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Boris Zhivotovsky
- Institute of Environmental Medicine, Division of Toxicology, Karolinska Institutet, Stockholm, Sweden
| | - Dan Grandér
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Lars Egevad
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Sten Nilsson
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Theocharis Panaretakis
- Department of Oncology-Pathology, Cancer Centrum Karolinska, Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
21
|
Effects of the multikinase inhibitors Sorafenib and Regorafenib in PTEN deficient neoplasias. Eur J Cancer 2016; 63:74-87. [DOI: 10.1016/j.ejca.2016.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 04/06/2016] [Accepted: 04/21/2016] [Indexed: 02/07/2023]
|
22
|
Kiprianova I, Remy J, Milosch N, Mohrenz IV, Seifert V, Aigner A, Kögel D. Sorafenib Sensitizes Glioma Cells to the BH3 Mimetic ABT-737 by Targeting MCL1 in a STAT3-Dependent Manner. Neoplasia 2016; 17:564-73. [PMID: 26297434 PMCID: PMC4547411 DOI: 10.1016/j.neo.2015.07.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/23/2015] [Accepted: 07/02/2015] [Indexed: 01/25/2023] Open
Abstract
The oncogenic transcription factor signal transducer and activator of transcription 3 (STAT3) is overactivated in malignant glioma and plays a key role in promoting cell survival, thereby increasing the acquired apoptosis resistance of these tumors. Here we investigated the STAT3/myeloid cell leukemia 1 (MCL1) signaling pathway as a target to overcome the resistance of glioma cells to the Bcl-2-inhibiting synthetic BH3 mimetic ABT-737. Stable lentiviral knockdown of MCL1 sensitized LN229 and U87 glioma cells to apoptotic cell death induced by single-agent treatment with ABT-737 which was associated with an early activation of DEVDase activity, cytochrome c release, and nuclear apoptosis. Similar sensitizing effects were observed when ABT-737 treatment was combined with the multikinase inhibitor sorafenib which effectively suppressed levels of phosphorylated STAT3 and MCL1 in MCL1-proficient LN229 and U87 glioma cells. In analogous fashion, these synergistic effects were observed when we combined ABT-737 with the STAT3 inhibitor WP-1066. Lentiviral knockdown of the activating transcription factor 5 combined with subsequent quantitative polymerase chain reaction analysis revealed that sorafenib-dependent suppression of MCL1 occurred at the transcriptional level but did not depend on activating transcription factor 5 which previously had been proposed to be essential for MCL1-dependent glioma cell survival. In contrast, the constitutively active STAT3 mutant STAT3-C was able to significantly enhance MCL1 levels under sorafenib treatment to retain cell survival. Collectively, these data demonstrate that sorafenib targets MCL1 in a STAT3-dependent manner, thereby sensitizing glioma cells to treatment with ABT-737. They also suggest that targeting STAT3 in combination with inducers of the intrinsic pathway of apoptosis may be a promising novel strategy for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Irina Kiprianova
- Experimental Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany
| | - Janina Remy
- Experimental Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany
| | - Nelli Milosch
- Experimental Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany
| | - Isabelle V Mohrenz
- Experimental Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany
| | - Volker Seifert
- Dept. of Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany
| | - Achim Aigner
- Rudolf-Boehm-Institute for Pharmacology and Toxicology, Clinical Pharmacology, University of Leipzig, Leipzig, Germany
| | - Donat Kögel
- Experimental Neurosurgery, Goethe University Hospital, Frankfurt am Main, Germany.
| |
Collapse
|
23
|
Toren P, Kim S, Johnson F, Zoubeidi A. Combined AKT and MEK Pathway Blockade in Pre-Clinical Models of Enzalutamide-Resistant Prostate Cancer. PLoS One 2016; 11:e0152861. [PMID: 27046225 PMCID: PMC4821639 DOI: 10.1371/journal.pone.0152861] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/20/2016] [Indexed: 11/25/2022] Open
Abstract
Despite recent improvements in patient outcomes using newer androgen receptor (AR) pathway inhibitors, treatment resistance in castrate resistant prostate cancer (CRPC) continues to remain a clinical problem. Co-targeting alternate resistance pathways are of significant interest to treat CRPC and delay the onset of resistance. Both the AKT and MEK signaling pathways become activated as prostate cancer develops resistance to AR-targeted therapies. This pre-clinical study explores co-targeting these pathways in AR-positive prostate cancer models. Using various in vitro models of prostate cancer disease states including androgen dependent (LNCaP), CRPC (V16D and 22RV1) and ENZ-resistant prostate cancer (MR49C and MR49F), we evaluate the relevance of targeting both AKT and MEK pathways. Our data reveal that AKT inhibition induces apoptosis and inhibits cell growth in PTEN null cell lines independently of their sensitivity to hormone therapy; however, AKT inhibition had no effect on the PTEN positive 22RV1 cell line. Interestingly, we found that MEK inhibition had greater effect on 22RV1 cells compared to LNCaP, V16D or ENZ-resistant cells MR49C and MR49F cells. In vitro, combination AKT and MEK blockade had evidence of synergy observed in some cell lines and assays, but this was not consistent across all results. In vivo, the combination of AKT and MEK inhibition resulted in more consistent tumor growth inhibition of MR49F xenografts and longer disease specific survival compared to AKT inhibitor monotherapy. As in our in vitro study, 22RV1 xenografts were more resistant to AKT inhibition while they were more sensitive to MEK inhibition. Our results suggest that targeting AKT and MEK in combination may be a valuable strategy in prostate cancer when both pathways are activated and further support the importance of characterizing the dominant oncogenic pathway in each patient's tumor in order to select optimal therapy.
Collapse
Affiliation(s)
- Paul Toren
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Soojin Kim
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Fraser Johnson
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Amina Zoubeidi
- The Vancouver Prostate Centre, Department of Urologic Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Pol J, Buqué A, Aranda F, Bloy N, Cremer I, Eggermont A, Erbs P, Fucikova J, Galon J, Limacher JM, Preville X, Sautès-Fridman C, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch-Oncolytic viruses and cancer therapy. Oncoimmunology 2016; 5:e1117740. [PMID: 27057469 PMCID: PMC4801444 DOI: 10.1080/2162402x.2015.1117740] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 02/06/2023] Open
Abstract
Oncolytic virotherapy relies on the administration of non-pathogenic viral strains that selectively infect and kill malignant cells while favoring the elicitation of a therapeutically relevant tumor-targeting immune response. During the past few years, great efforts have been dedicated to the development of oncolytic viruses with improved specificity and potency. Such an intense wave of investigation has culminated this year in the regulatory approval by the US Food and Drug Administration (FDA) of a genetically engineered oncolytic viral strain for use in melanoma patients. Here, we summarize recent preclinical and clinical advances in oncolytic virotherapy.
Collapse
Affiliation(s)
- Jonathan Pol
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Aitziber Buqué
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d’Investigacions Biomédiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Norma Bloy
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | | | | | - Jitka Fucikova
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers, Paris, France
| | | | | | - Catherine Sautès-Fridman
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 13, Center de Recherche des Cordeliers, Paris, France
| | - Radek Spisek
- Sotio, Prague, Czech Republic
- Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France
- INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
- Gustave Roussy Cancer Campus, Villejuif, France
| |
Collapse
|
25
|
Cancer-Associated Fibroblasts: Their Characteristics and Their Roles in Tumor Growth. Cancers (Basel) 2015; 7:2443-58. [PMID: 26690480 PMCID: PMC4695902 DOI: 10.3390/cancers7040902] [Citation(s) in RCA: 586] [Impact Index Per Article: 58.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 11/17/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022] Open
Abstract
Cancer tissues are composed of cancer cells and the surrounding stromal cells (e.g., fibroblasts, vascular endothelial cells, and immune cells), in addition to the extracellular matrix. Most studies investigating carcinogenesis and the progression, invasion, metastasis, and angiogenesis of cancer have focused on alterations in cancer cells, including genetic and epigenetic changes. Recently, interactions between cancer cells and the stroma have attracted considerable attention, and increasing evidence has accumulated on this. Several researchers have gradually clarified the origins, features, and roles of cancer-associated fibroblasts (CAFs), a major component of the cancer stroma. CAFs function in a similar manner to myofibroblasts during wound healing. We previously reported the relationship between CAFs and angiogenesis. Interleukin-6 (IL-6), a multifunctional cytokine, plays a central role in regulating inflammatory and immune responses, and important roles in the progression, including proliferation, migration, and angiogenesis, of several cancers. We showed that CAFs are an important IL-6 source and that anti-IL-6 receptor antibody suppressed angiogenesis and inhibited tumor-stroma interactions. Furthermore, CAFs contribute to drug-resistance acquisition in cancer cells. The interaction between cancer cells and the stroma could be a potential target for anti-cancer therapy.
Collapse
|
26
|
Gunaydin G, Kesikli SA, Guc D. Cancer associated fibroblasts have phenotypic and functional characteristics similar to the fibrocytes that represent a novel MDSC subset. Oncoimmunology 2015; 4:e1034918. [PMID: 26405600 DOI: 10.1080/2162402x.2015.1034918] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/19/2015] [Accepted: 03/21/2015] [Indexed: 12/17/2022] Open
Abstract
Circulating fibrocytes were reported to represent a novel myeloid-derived suppressor cell (MDSC) subset and they were also proposed to be involved in the tumor immune escape. This novel fibrocyte subset had a surface phenotype resembling non-monocytic MDSCs (CD14-CD11chiCD123-) and exhibited immunomodulatory roles. Most effector functions of fibrocytes (circulating fibroblast-progenitors) are accomplished as tissue fibroblasts, likewise in the tumor microenvironment. Therefore, fibroblasts at tumor tissues should be evaluated whether they display similar molecular/gene expression patterns and functional roles to the blood-borne fibrocytes. A chemically induced rat breast carcinogenesis model was utilized to obtain cancer associated fibroblasts (CAFs). CAFs and normal tissue fibroblasts (NFs) were isolated from cancerous and healthy breast tissues, respectively, using a previously described enzymatic protocol. Both CAFs and NFs were analyzed for cell surface phenotypes by flow cytometry and for gene expression profiles by gene set enrichment analysis (GSEA). PBMCs were cocultured with either NFs or CAFs and proliferations of PBMCs were assessed by CFSE assays. Morphological analyses were performed by immunocytochemistry stainings with vimentin. CAFs were spindle shaped cells unlike their blood-borne counterparts. They did not express CD80 and their MHC-II expression was lower than NFs. Although CAFs expressed the myeloid marker CD11b/c, its expression was lower than that on the circulating fibrocytes. CAFs did not express granulocytic/neutrophilic markers and they seemed to have developed in an environment containing THELPER2-like cytokines. They also showed immunosuppressive effects similar to their blood-borne counterparts. In summary, CAFs showed similar phenotypic and functional characteristics to the circulating fibrocytes that were reported to represent a unique MDSC subset.
Collapse
Affiliation(s)
- Gurcan Gunaydin
- Department of Basic Oncology; Hacettepe University Cancer Institute ; Sihhiye, Ankara, Turkey
| | - S Altug Kesikli
- Department of Basic Oncology; Hacettepe University Cancer Institute ; Sihhiye, Ankara, Turkey
| | - Dicle Guc
- Department of Basic Oncology; Hacettepe University Cancer Institute ; Sihhiye, Ankara, Turkey
| |
Collapse
|
27
|
Fan Z, Yu H, Cui N, Kong X, Liu X, Chang Y, Wu Y, Sun L, Wang G. ABT737 enhances cholangiocarcinoma sensitivity to cisplatin through regulation of mitochondrial dynamics. Exp Cell Res 2015; 335:68-81. [PMID: 25936772 DOI: 10.1016/j.yexcr.2015.04.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 03/18/2015] [Accepted: 04/22/2015] [Indexed: 12/17/2022]
Abstract
Cholangiocarcinoma responses weakly to cisplatin. Mitochondrial dynamics participate in the response to various stresses, and mainly involve mitophagy and mitochondrial fusion and fission. Bcl-2 family proteins play critical roles in orchestrating mitochondrial dynamics, and are involved in the resistance to cisplatin. Here we reported that ABT737, combined with cisplatin, can promote cholangiocarcinoma cells to undergo apoptosis. We found that the combined treatment decreased the Mcl-1 pro-survival form and increased Bak. Cells undergoing cisplatin treatment showed hyperfused mitochondria, whereas fragmentation was dominant in the mitochondria of cells exposed to the combined treatment, with higher Fis1 levels, decreased Mfn2 and OPA1 levels, increased ratio of Drp1 60kD to 80kD form, and more Drp1 located on mitochondria. More p62 aggregates were observed in cells with fragmented mitochondria, and they gradually translocated to mitochondria. Mitophagy was induced by the combined treatment. Knockdown p62 decreased the Drp1 ratio, increased Tom20, and increased cell viability. Our data indicated that mitochondrial dynamics play an important role in the response of cholangiocarcinoma to cisplatin. ABT737 might enhance cholangiocarcinoma sensitivity to cisplatin through regulation of mitochondrial dynamics and the balance within Bcl-2 family proteins. Furthermore, p62 seems to be critical in the regulation of mitochondrial dynamics.
Collapse
Affiliation(s)
- Zhongqi Fan
- Department of Hepatobiliary & Pancreas Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Huimei Yu
- Department of Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Ni Cui
- Bethune Medical College, Jilin University, Changchun, Jilin 130021, China
| | - Xianggui Kong
- State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin 130033, China
| | - Xiaomin Liu
- State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin 130033, China
| | - Yulei Chang
- State Key Laboratory of Luminescence and Applications, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun, Jilin 130033, China
| | - Yao Wu
- Department of Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Liankun Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Guangyi Wang
- Department of Hepatobiliary & Pancreas Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
28
|
Obrist F, Manic G, Kroemer G, Vitale I, Galluzzi L. Trial Watch: Proteasomal inhibitors for anticancer therapy. Mol Cell Oncol 2015; 2:e974463. [PMID: 27308423 PMCID: PMC4904962 DOI: 10.4161/23723556.2014.974463] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 09/15/2014] [Accepted: 09/17/2014] [Indexed: 01/12/2023]
Abstract
The so-called "ubiquitin-proteasome system" (UPS) is a multicomponent molecular apparatus that catalyzes the covalent attachment of several copies of the small protein ubiquitin to other proteins that are generally (but not always) destined to proteasomal degradation. This enzymatic cascade is crucial for the maintenance of intracellular protein homeostasis (both in physiological conditions and in the course of adaptive stress responses), and regulates a wide array of signaling pathways. In line with this notion, defects in the UPS have been associated with aging as well as with several pathological conditions including cardiac, neurodegenerative, and neoplastic disorders. As transformed cells often experience a constant state of stress (as a result of the hyperactivation of oncogenic signaling pathways and/or adverse microenvironmental conditions), their survival and proliferation are highly dependent on the integrity of the UPS. This rationale has driven an intense wave of preclinical and clinical investigation culminating in 2003 with the approval of the proteasomal inhibitor bortezomib by the US Food and Drug Administration for use in multiple myeloma patients. Another proteasomal inhibitor, carfilzomib, is now licensed by international regulatory agencies for use in multiple myeloma patients, and the approved indications for bortezomib have been extended to mantle cell lymphoma. This said, the clinical activity of bortezomib and carfilzomib is often limited by off-target effects, innate/acquired resistance, and the absence of validated predictive biomarkers. Moreover, the antineoplastic activity of proteasome inhibitors against solid tumors is poor. In this Trial Watch we discuss the contribution of the UPS to oncogenesis and tumor progression and summarize the design and/or results of recent clinical studies evaluating the therapeutic profile of proteasome inhibitors in cancer patients.
Collapse
Affiliation(s)
- Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | | | - Guido Kroemer
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- Department of Biology, University of Rome “Tor Vergata”
| | - Lorenzo Galluzzi
- INSERM, U1138; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
29
|
Nouraee N, Mowla SJ, Calin GA. Tracking miRNAs' footprints in tumor-microenvironment interactions: Insights and implications for targeted cancer therapy. Genes Chromosomes Cancer 2015; 54:335-52. [PMID: 25832733 DOI: 10.1002/gcc.22244] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 01/16/2015] [Accepted: 01/17/2015] [Indexed: 12/16/2022] Open
Abstract
In the past decades, cancer medicine studies have mainly focused on tumor cell biology as the main promoter of solid tumor progression. However, tumor biology does not explain the intertwinement and ambiguity of the tumors' territory. Recently, the approach of understanding cancer has shifted from investigating the biology of tumor cells to studying the microenvironment surrounding them. MicroRNAs (miRNAs), which play a role in exploiting indigenous stromal cells and are components that cooperate and produce a favorable microenvironment for progressive tumor formation, have been implicated in numerous processes essential for tumor initiation and growth. Understanding the mechanisms underlying interactions between tumor cells and their adjacent environment holds many promises for the future of cancer-targeted therapies. Herein, we provide a step-by-step account of miRNA involvement in tumor-microenvironment interactions as the micromediators of tumor cell and stroma communications. We also focus on the clinical challenges in using miRNAs tof overcome therapy resistance mechanisms and tumor heterogeneity bias in cancer therapy.
Collapse
Affiliation(s)
- Nazila Nouraee
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | |
Collapse
|
30
|
Pol J, Vacchelli E, Aranda F, Castoldi F, Eggermont A, Cremer I, Sautès-Fridman C, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology 2015; 4:e1008866. [PMID: 26137404 DOI: 10.1080/2162402x.2015.1008866] [Citation(s) in RCA: 228] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 01/14/2015] [Indexed: 02/06/2023] Open
Abstract
The term "immunogenic cell death" (ICD) is now employed to indicate a functionally peculiar form of apoptosis that is sufficient for immunocompetent hosts to mount an adaptive immune response against dead cell-associated antigens. Several drugs have been ascribed with the ability to provoke ICD when employed as standalone therapeutic interventions. These include various chemotherapeutics routinely employed in the clinic (e.g., doxorubicin, epirubicin, idarubicin, mitoxantrone, bleomycin, bortezomib, cyclophosphamide and oxaliplatin) as well as some anticancer agents that are still under preclinical or clinical development (e.g., some microtubular inhibitors of the epothilone family). In addition, a few drugs are able to convert otherwise non-immunogenic instances of cell death into bona fide ICD, and may therefore be employed as chemotherapeutic adjuvants within combinatorial regimens. This is the case of cardiac glycosides, like digoxin and digitoxin, and zoledronic acid. Here, we discuss recent developments on anticancer chemotherapy based on ICD inducers.
Collapse
Key Words
- ALL, acute lymphoblastic leukemia
- AML, acute myeloid leukemia
- CML, chronic myeloid leukemia
- DAMP, damage-associated molecular pattern
- EGFR, epidermal growth factor receptor
- EOX, epirubicin plus oxaliplatin plus capecitabine
- ER, endoplasmic reticulum
- FDA, Food and Drug Administration
- FOLFIRINOX, folinic acid plus 5-fluorouracil plus irinotecan plus oxaliplatin
- FOLFOX, folinic acid plus 5-fluorouracil plus oxaliplatin
- GEMOX, gemcitabine plus oxaliplatin
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- HCC, hepatocellular carcinoma
- ICD, immunogenic cell death
- MM, multiple myeloma
- NHL, non-Hodgkin's lymphoma
- NSCLC, non-small cell lung carcinoma
- TACE, transcatheter arterial chemoembolization
- XELOX, capecitabine plus oxaliplatin
- antigen-presenting cell
- autophagy
- damage-associated molecular pattern
- dendritic cell
- endoplasmic reticulum stress
- mAb, monoclonal antibody
- type I interferon
Collapse
Affiliation(s)
- Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France
| | - Erika Vacchelli
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France
| | - Fernando Aranda
- Group of Immune receptors of the Innate and Adaptive System, Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS)
| | - Francesca Castoldi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Faculté de Medicine; Université Paris Sud/Paris XI ; Le Kremlin-Bicêtre, France ; Sotio a.c. ; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM, U1138 ; Paris, France ; Equipe 13, Center de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, U1138 ; Paris, France ; Equipe 13, Center de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Jitka Fucikova
- Sotio a.c. ; Prague, Czech Republic ; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138 ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| | - Radek Spisek
- Sotio a.c. ; Prague, Czech Republic ; Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University ; Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; INSERM , U970 ; Paris, France ; Paris-Cardiovascular Research Center (PARCC) ; Paris, France ; Service d'Immunologie Biologique, Hôpital Européen Georges Pompidou (HEGP); AP-HP ; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015; CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou; AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
31
|
Poovassery JS, Kang JC, Kim D, Ober RJ, Ward ES. Antibody targeting of HER2/HER3 signaling overcomes heregulin-induced resistance to PI3K inhibition in prostate cancer. Int J Cancer 2014; 137:267-77. [PMID: 25471734 DOI: 10.1002/ijc.29378] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 11/18/2014] [Indexed: 12/11/2022]
Abstract
Dysregulated expression and/or mutations of the various components of the phosphoinositide 3-kinase (PI3K)/Akt pathway occur with high frequency in prostate cancer and are associated with the development and progression of castration resistant tumors. However, small molecule kinase inhibitors that target this signaling pathway have limited efficacy in inhibiting tumor growth, primarily due to compensatory survival signals through receptor tyrosine kinases (RTKs). Although members of the epidermal growth factor receptor (EGFR), or HER, family of RTKs are strongly implicated in the development and progression of prostate cancer, targeting individual members of this family such as EGFR or HER2 has resulted in limited success in clinical trials. Multiple studies indicate a critical role for HER3 in the development of resistance against both HER-targeted therapies and PI3K/Akt pathway inhibitors. In this study, we found that the growth inhibitory effect of GDC-0941, a class I PI3K inhibitor, is markedly reduced in the presence of heregulin. Interestingly, this effect is more pronounced in cells lacking phosphatase and tensin homolog function. Heregulin-mediated resistance to GDC-0941 is associated with reactivation of Akt downstream of HER3 phosphorylation. Importantly, combined blockade of HER2 and HER3 signaling by an anti-HER2/HER3 bispecific antibody or a mixture of anti-HER2 and anti-HER3 antibodies restores sensitivity to GDC-0941 in heregulin-treated androgen-dependent and -independent prostate cancer cells. These studies indicate that the combination of PI3K inhibitors with HER2/HER3 targeting antibodies may constitute a promising therapeutic strategy for prostate cancer.
Collapse
Affiliation(s)
| | - Jeffrey C Kang
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, College Station, TX
| | - Dongyoung Kim
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, College Station, TX.,Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - Raimund J Ober
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, College Station, TX.,Department of Biomedical Engineering, Texas A&M University, College Station, TX
| | - E Sally Ward
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College of Medicine, College Station, TX.,Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX
| |
Collapse
|
32
|
Differential sensitivity of prostate tumor derived endothelial cells to sorafenib and sunitinib. BMC Cancer 2014; 14:939. [PMID: 25494980 PMCID: PMC4295225 DOI: 10.1186/1471-2407-14-939] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 12/04/2014] [Indexed: 11/26/2022] Open
Abstract
Background Prostate cancer is the second leading cause of male cancer death in developed countries. Although the role of angiogenesis in its progression is well established, the efficacy of anti-angiogenic therapy is not clearly proved. Whether this could depend on differential responses between tumor and normal endothelial cells has not been tested. Methods We isolated and characterized three lines of endothelial cells from prostate cancer and we tested the effect of Sunitinib and Sorafenib, and the combined treatment with the anti-androgen Casodex, on their angiogenic functions. Results Endothelial cells isolated from prostate tumors showed angiogenic properties and expression of androgen and vascular endothelial cell growth factor receptors. Sunitinib affected their proliferation, survival and motility while Sorafenib only showed a minor effect. At variance, Sunitinib and Sorafenib showed similar cytotoxic and anti-angiogenic effects on normal endothelial cells. Sorafenib and Sunitinib inhibited vascular endothelial cell growth factor receptor2 phosphorylation of prostate cancer endothelial cells, while they differentially modulated Akt phosphorylation as no inhibitory effect of Sorafenib was observed on Akt activation. The combined treatment of Casodex reverted the observed resistance to Sorafenib both on cell viability and on Akt activation, whereas it did not modify the response to Sunitinib. Conclusions Our study demonstrates a resistant behavior of endothelial cells isolated from prostate cancer to Sorafenib, but not Sunitinib. Moreover, it shows the benefit of a multi-target therapy combining anti-angiogenic and anti-hormonal drugs to overcome resistance. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-939) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Manic G, Obrist F, Kroemer G, Vitale I, Galluzzi L. Chloroquine and hydroxychloroquine for cancer therapy. Mol Cell Oncol 2014; 1:e29911. [PMID: 27308318 PMCID: PMC4905171 DOI: 10.4161/mco.29911] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/16/2014] [Indexed: 02/06/2023]
Abstract
Macroautophagy (herein referred to as autophagy) is a highly conserved mechanism for the lysosomal degradation of cytoplasmic components. Autophagy is critical for the maintenance of intracellular homeostasis, both in baseline conditions and in the context of adaptive responses to stress. In line with this notion, defects in the autophagic machinery have been etiologically associated with various human disorders including infectious, inflammatory and neoplastic conditions. Once tumors are established, however, autophagy sustains the survival of malignant cells, hence representing an appealing target for the design of novel anticancer regimens. Accordingly, inhibitors of autophagy including chloroquine and hydroxychloroquine have been shown to mediate substantial antineoplastic effects in preclinical models, especially when combined with chemo- or radiotherapeutic interventions. The pharmacological profile of chloroquine and hydroxychloroquine, however, appear to involve mechanisms other than autophagy inhibition. Here, we discuss the dual role of autophagy in oncogenesis and tumor progression, and summarize the results or design of clinical studies recently completed or initiated to evaluate the therapeutic activity of chloroquine derivatives in cancer patients.
Collapse
Affiliation(s)
| | - Florine Obrist
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France; INSERM, UMRS1138; Villejuif, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | - Guido Kroemer
- INSERM, UMRS1138; Villejuif, France; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Paris, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Lorenzo Galluzzi
- Regina Elena National Cancer Institute; Rome, Italy; Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
34
|
Elsner A, Lange F, Fitzner B, Heuschkel M, Krause BJ, Jaster R. Distinct antifibrogenic effects of erlotinib, sunitinib and sorafenib on rat pancreatic stellate cells. World J Gastroenterol 2014; 20:7914-7925. [PMID: 24976727 PMCID: PMC4069318 DOI: 10.3748/wjg.v20.i24.7914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/09/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To study if three clinically available small molecule kinase inhibitors (SMI), erlotinib, sunitinib and sorafenib, exert antifibrogenic effects on pancreatic stellate cells (PSC) and analyze the basis of their action.
METHODS: Cultured rat PSC were exposed to SMI. Cell proliferation and viability were assessed employing 5-bromo-2’-deoxyuridine incorporation assay and flow cytometry, respectively. 2-Deoxy-2-[18F] fluoroglucose (18F-FDG) uptake was measured to study metabolic activity. Exhibition of the myofibroblastic PSC phenotype was monitored by immunofluorescence analysis of α-smooth muscle actin (α-SMA) expression. Levels of mRNA were determined by real-time PCR, while protein expression and phosphorylation were analyzed by immunoblotting. Transforming growth factor-β1 (TGF-β1) levels in culture supernatants were quantified by ELISA.
RESULTS: All three SMI inhibited cell proliferation and 18F-FDG uptake in a dose-dependent manner and without significant cytotoxic effects. Furthermore, additive effects of the drugs were observed. Immunoblot analysis showed that sorafenib and sunitib, but not erlotinib, efficiently blocked activation of the AKT pathway, while all three drugs displayed little effect on phosphorylation of ERK1/2. Cells treated with sorafenib or sunitinib expressed less interleukin-6 mRNA as well as less collagen type 1 mRNA and protein. Sorafenib was the only drug that also upregulated the expression of matrix metalloproteinase-2 and reduced the secretion of TGF-β1 protein. All three drugs showed insignificant or discordant effects on the mRNA and protein levels of α-SMA.
CONCLUSION: The tested SMI, especially sorafenib, exert inhibitory effects on activated PSC, which should be further evaluated in preclinical studies.
Collapse
|
35
|
Weigel KJ, Jakimenko A, Conti BA, Chapman SE, Kaliney WJ, Leevy WM, Champion MM, Schafer ZT. CAF-Secreted IGFBPs Regulate Breast Cancer Cell Anoikis. Mol Cancer Res 2014; 12:855-66. [DOI: 10.1158/1541-7786.mcr-14-0090] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. Oncoimmunology 2014; 3:e28344. [PMID: 25050207 PMCID: PMC4063152 DOI: 10.4161/onci.28344] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 02/24/2014] [Indexed: 12/19/2022] Open
Abstract
The expression "adoptive cell transfer" (ACT) is commonly employed to indicate an immunotherapeutic regimen involving the isolation of autologous blood-borne or tumor-infiltrating lymphocytes, their selection/expansion/activation ex vivo, and their reinfusion into the patient, most often in the context of lymphodepleting pre-conditioning and in combination with immunostimulatory treatments. Optionally, the cellular material for ACT is genetically manipulated before expansion to (1) target specific tumor-associated antigens; (2) endogenously express immunostimulatory molecules; and/or (3) persist for long periods upon reinfusion. Consistent efforts have been dedicated at the amelioration of this immunotherapeutic regimen throughout the past decade, resulting in the establishment of ever more efficient and safer ACT protocols. Accordingly, the number of clinical trials testing ACT in oncological indications does not cease to increase. In this Trial Watch, we summarize recent developments in this exciting area of research, covering both high-impact studies that have been published during the last 12 months and clinical trials that have been launched in the same period to evaluate the safety and therapeutic potential of ACT in cancer patients.
Collapse
Affiliation(s)
- Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- INSERM, UMRS1138; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 15, Centre de Recherche des Cordeliers; Paris, France
| | - Wolf Hervé Fridman
- INSERM, UMRS1138; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Isabelle Cremer
- INSERM, UMRS1138; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; Equipe 13, Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Eric Tartour
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; INSERM, U970; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
37
|
Bruzzese F, Hägglöf C, Leone A, Sjöberg E, Roca MS, Kiflemariam S, Sjöblom T, Hammarsten P, Egevad L, Bergh A, Ostman A, Budillon A, Augsten M. Local and systemic protumorigenic effects of cancer-associated fibroblast-derived GDF15. Cancer Res 2014; 74:3408-17. [PMID: 24780757 DOI: 10.1158/0008-5472.can-13-2259] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tumor stroma is vital to tumor development, progression, and metastasis. Cancer-associated fibroblasts (CAF) are among the abundant cell types in the tumor stroma, but the range of their contributions to cancer pathogenicity has yet to be fully understood. Here, we report a critical role for upregulation of the TGFβ/BMP family member GDF15 (MIC-1) in tumor stroma. GDF15 was found upregulated in situ and in primary cultures of CAF from prostate cancer. Ectopic expression of GDF15 in fibroblasts produced prominent paracrine effects on prostate cancer cell migration, invasion, and tumor growth. Notably, GDF15-expressing fibroblasts exerted systemic in vivo effects on the outgrowth of distant and otherwise indolent prostate cancer cells. Our findings identify tumor stromal cells as a novel source of GDF15 in human prostate cancer and illustrate a systemic mechanism of cancer progression driven by the tumor microenvironment. Further, they provide a functional basis to understand GDF15 as a biomarker of poor prognosis and a candidate therapeutic target in prostate cancer.
Collapse
Affiliation(s)
- Francesca Bruzzese
- Authors' Affiliations: Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"-IRCCS, Naples, Italy
| | | | - Alessandra Leone
- Authors' Affiliations: Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"-IRCCS, Naples, Italy
| | - Elin Sjöberg
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Maria Serena Roca
- Authors' Affiliations: Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"-IRCCS, Naples, Italy
| | - Sara Kiflemariam
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala; and
| | - Tobias Sjöblom
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala; and
| | - Peter Hammarsten
- Department of Medical Biosciences, Pathology, Umeå University, Umeå
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Pathology, Umeå University, Umeå
| | - Arne Ostman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Alfredo Budillon
- Authors' Affiliations: Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale"-IRCCS, Naples, Italy
| | - Martin Augsten
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
38
|
Vacchelli E, Aranda F, Eggermont A, Galon J, Sautès-Fridman C, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2014; 3:e27878. [PMID: 24800173 PMCID: PMC4008470 DOI: 10.4161/onci.27878] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 12/22/2022] Open
Abstract
Accumulating evidence suggests that the clinical efficacy of selected anticancer drugs, including conventional chemotherapeutics as well as targeted anticancer agents, originates (at least in part) from their ability to elicit a novel or reinstate a pre-existing tumor-specific immune response. One of the mechanisms whereby chemotherapy can stimulate the immune system to recognize and destroy malignant cells is commonly known as immunogenic cell death (ICD). Cancer cells succumbing to ICD are de facto converted into an anticancer vaccine and as such elicit an adaptive immune response. Several common chemotherapeutics share the ability of triggering ICD, as demonstrated in vaccination experiments relying on immunocompetent mice and syngeneic cancer cells. A large number of ongoing clinical trials involve such ICD inducers, often (but not always) as they are part of the gold standard therapeutic approach against specific neoplasms. In this Trial Watch, we summarize the latest advances on the use of cyclophosphamide, doxorubicin, epirubicin, oxaliplatin, and mitoxantrone in cancer patients, discussing high-impact studies that have been published during the last 13 months as well as clinical trials that have been initiated in the same period to assess the antineoplastic profile of these immunogenic drugs as off-label therapeutic interventions.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | - Fernando Aranda
- Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Université Paris-Sud/Paris XI; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| | | | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France ; Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- Université Pierre et Marie Curie/Paris VI; Paris, France ; INSERM, UMRS1138; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France ; INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy; Villejuif, France ; INSERM, U848; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|
39
|
Gumulec J, Balvan J, Sztalmachova M, Raudenska M, Dvorakova V, Knopfova L, Polanska H, Hudcova K, Ruttkay-Nedecky B, Babula P, Adam V, Kizek R, Stiborova M, Masarik M. Cisplatin-resistant prostate cancer model: Differences in antioxidant system, apoptosis and cell cycle. Int J Oncol 2013; 44:923-33. [PMID: 24366574 DOI: 10.3892/ijo.2013.2223] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 11/11/2013] [Indexed: 11/06/2022] Open
Abstract
Differences in the antioxidant system, apoptotic mechanism and in cell cycle between prostatic cell lines could partially elucidate the development of cisplatin resistance. The aim of this study was to identify the most characteristic parameter for a particular cell line and/or a particular cisplatin treatment using a general regression model and to assess whether it is possible to use measured parameters as markers of cisplatin resistance. This study integrates the results of viability, antioxidant, flow cytometric and quantitative PCR assays in order to characterize the resistance of prostate cancer to cisplatin. Cell growth using metabolic- (MTT) and impedance-based assays, the expression of key cell death signaling proteins (p53, Bax and Bcl-2), cell cycle, activity of antioxidant system-related proteins (superoxide dismutase, glutathione peroxidase, glutathione reductase and metallothionein) and free radical scavenging capacity assays [free radicals (FR), ferric reducing antioxidant power (FRAP), ABTS] were analyzed in the cell lines 22Rv1, PC-3 and PNT1A with respect to rising concentrations (0-150 µM) and different length of cisplatin treatment (12-72 h). The non-functional-p53 PC-3 cell line showed decreased BAX (p<0.05) and, in contrast to PNT1A and 22Rv1, no cisplatin-induced effects on cell cycle. All cell lines showed increasing levels of free radical scavenging activity by ABTS, FRAP and FR assays in a time- and dose-dependent manner (r>0.76 at p<0.001 for ABTS, FRAP and FR at p<0.001). PC-3 showed increased (p<0.05) levels of free radical scavenging activity by ABTS and FR methods. These findings, together with significantly elevated MT, decreased p53 and Bax indicate PC-3 to be cisplatin-resistant. The differences in the antioxidant system and apoptotic mechanisms in PC-3 cells may elucidate the development of cisplatin resistance and indicate that this cell line may be further studied as a model of cytostatic resistance.
Collapse
Affiliation(s)
- Jaromir Gumulec
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Jan Balvan
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Marketa Sztalmachova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Veronika Dvorakova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Lucia Knopfova
- Department of Experimental Biology, Faculty of Science, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Hana Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | - Kristyna Hudcova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| | | | - Petr Babula
- Department of Natural Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, CZ-612 42 Brno, Czech Republic
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-613 00 Brno, Czech Republic
| | - Rene Kizek
- Department of Chemistry and Biochemistry, Mendel University in Brno, CZ-613 00 Brno, Czech Republic
| | - Marie Stiborova
- Department of Biochemistry, Faculty of Science, Charles University, CZ-128 40 Prague 2, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, CZ-625 00 Brno, Czech Republic
| |
Collapse
|
40
|
Onnis B, Fer N, Rapisarda A, Perez VS, Melillo G. Autocrine production of IL-11 mediates tumorigenicity in hypoxic cancer cells. J Clin Invest 2013; 123:1615-29. [PMID: 23549086 PMCID: PMC3613900 DOI: 10.1172/jci59623] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 01/24/2013] [Indexed: 12/22/2022] Open
Abstract
IL-11 and its receptor, IL-11Ra, are expressed in human cancers; however, the functional role of IL-11 in tumor progression is not known. We found that IL11 is a hypoxia-inducible, VHL-regulated gene in human cancer cells and that expression of IL11 mRNA was dependent, at least in part, on HIF-1. A cooperative interaction between HIF-1 and AP-1 mediated transcriptional activation of the IL11 promoter. Additionally, we found that human cancer cells expressed a functional IL-11Ra subunit, which triggered signal transduction either by exogenous recombinant human IL-11 or by autocrine production of IL-11 in cells cultured under hypoxic conditions. Silencing of IL11 dramatically abrogated the ability of hypoxia to increase anchorage-independent growth and significantly reduced tumor growth in xenograft models. Notably, these results were phenocopied by partial knockdown of STAT1 in a human prostate cancer cell line (PC3), suggesting that this pathway may play an important role in mediating the effects of IL-11 under hypoxic conditions. In conclusion, these results identify IL11 as an oxygen- and VHL-regulated gene and provide evidence of a pathway "hijacked" by hypoxic cancer cells that may contribute to tumor progression.
Collapse
Affiliation(s)
- Barbara Onnis
- Developmental Therapeutics Program and
SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Nicole Fer
- Developmental Therapeutics Program and
SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Annamaria Rapisarda
- Developmental Therapeutics Program and
SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Victor S. Perez
- Developmental Therapeutics Program and
SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Giovanni Melillo
- Developmental Therapeutics Program and
SAIC-Frederick Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| |
Collapse
|
41
|
Adenovirus-mediated sensitization to the cytotoxic drugs docetaxel and mitoxantrone is dependent on regulatory domains in the E1ACR1 gene-region. PLoS One 2012; 7:e46617. [PMID: 23056370 PMCID: PMC3463540 DOI: 10.1371/journal.pone.0046617] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2012] [Accepted: 09/05/2012] [Indexed: 12/23/2022] Open
Abstract
Oncolytic adenoviruses have shown promising efficacy in clinical trials targeting prostate cancers that frequently develop resistance to all current therapies. The replication-selective mutants AdΔΔ and dl922–947, defective in pRb-binding, have been demonstrated to synergise with the current standard of care, mitoxantrone and docetaxel, in prostate cancer models. While expression of the early viral E1A gene is essential for the enhanced cell killing, the specific E1A-regions required for the effects are unknown. Here, we demonstrate that replicating mutants deleted in small E1A-domains, binding pRb (dl1108), p300/CBP (dl1104) and p400/TRRAP or p21 (dl1102) sensitize human prostate cancer cells (PC-3, DU145, 22Rv1) to mitoxantrone and docetaxel. Through generation of non-replicating mutants, we demonstrate that the small E1A12S protein is sufficient to potently sensitize all prostate cancer cells to the drugs even in the absence of viral replication and the E1A transactivating domain, conserved region (CR) 3. Furthermore, the p300/CBP-binding domain in E1ACR1 is essential for drug-sensitisation in the absence (AdE1A1104) but not in the presence of the E1ACR3 (dl1104) domain. AdE1A1104 also failed to increase apoptosis and accumulation of cells in G2/M. All E1AΔCR2 mutants (AdE1A1108, dl922–947) and AdE1A1102 or dl1102 enhance cell killing to the same degree as wild type virus. In PC-3 xenografts in vivo the dl1102 mutant significantly prolongs time to tumor progression that is further enhanced in combination with docetaxel. Neither dl1102 nor dl1104 replicates in normal human epithelial cells (NHBE). These findings suggest that additional E1A-deletions might be included when developing more potent replication-selective oncolytic viruses, such as the AdΔCR2-mutants, to further enhance potency through synergistic cell killing in combination with current chemotherapeutics.
Collapse
|
42
|
Parray A, Siddique HR, Nanda S, Konety BR, Saleem M. Castration-resistant prostate cancer: potential targets and therapies. Biologics 2012; 6:267-76. [PMID: 22956858 PMCID: PMC3430091 DOI: 10.2147/btt.s23954] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The treatment landscape for patients with castration-resistant prostate cancer (CRPC) is undergoing significant changes with the advent of new therapies and multidisciplinary efforts by scientists and clinicians. As activation of multiple molecular pathways in the neoplastic prostate makes it impossible for single-target drugs to be completely effective in treating CRPC, this has led to combination therapy strategy, where several molecules involved in tumor growth and disease progression are targeted by a therapeutic regimen. In the present review, we provide an update on the molecular pathways that play an important role in the pathogenesis of CRPC and discuss the current wave of new treatments to combat this lethal disease.
Collapse
Affiliation(s)
- Aijaz Parray
- Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, TX
| | - Hifzur R Siddique
- Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, TX
| | - Sanjeev Nanda
- Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, TX
- Department of Internal Medicine, Mayo Clinic Health Systems, Austin, TX
| | | | - Mohammad Saleem
- Molecular Chemoprevention and Therapeutics, The Hormel Institute, University of Minnesota, Austin, TX
- Department of Urology, University of Minnesota, Minneapolis
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|