1
|
Chen MF, Chen YY, Chen WC, Hsieh CC. The relationship of nutritional status with anticancer immunity and its prognostic value for head and neck cancer. Mol Carcinog 2023; 62:1388-1398. [PMID: 37265427 DOI: 10.1002/mc.23584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/27/2023] [Accepted: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Malnutrition has been reported to be associated with reduced survival and deficient anticancer immunity, and undernourishment is a frequent comorbidity in head and neck cancer (HNC) patients. In this study, we evaluated the relationship between nutritional status and immunologic factors, and its prognostic value for HNC. We retrospectively reviewed 212 HNC patients who had undergone a nutrition evaluation based on the Patient-Generated Subjective Global Assessment (PG-SGA) and curative radiotherapy (RT). The role of nutritional status in the prognosis of HNC and its correlation with anticancer immune response was assessed in HNC patients, and in the 4-nitroquinoline 1-oxide (4NQO)-induced tongue tumor animal model. Our data revealed that malnutrition (high PG-SGA scores) was significantly associated with more advanced disease, lower body mass index, lower RT completion rates, and reduced survival. Patients in the group with high PG-SGA scores had a higher neutrophil-to-lymphocyte ratio, higher proportion of myeloid-derived suppressor cells (MDSCs), and elevated IL-6 levels in the peripheral circulation. Patients with increased PG-SGA scores following treatment were more likely to developing locoregional failure. In the 4NQO-induced tumor model, nutritional supplementation decreased the rate of invasive tumor formation and attenuated the immune-suppressive microenvironment. Following ectopic tumor implantation in an immunocompetent host, nutrition supplements decreased tumor growth in association with attenuated MDSC recruitment and lower IL-6 expression. In conclusion, malnutrition by PG-SGA was associated with poor prognosis in HNC patients. Based on the data of HNC patients and the 4NQO-tumor model, adequate nutritional supplementation might improve the prognosis associated with augmented anticancer immunity.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Linko, Taoyuan, Taiwan
| | - Yu-Yen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Wen-Cheng Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| | - Ching-Chuan Hsieh
- Department of General Surgery, Chang Gung Memorial Hospital at Chiayi, Chiayi, Taiwan
| |
Collapse
|
2
|
Weng YS, Chiang IT, Tsai JJ, Liu YC, Hsu FT. Lenvatinib Synergistically Promotes Radiation Therapy in Hepatocellular Carcinoma by Inhibiting Src/STAT3/NF-κB-Mediated Epithelial-Mesenchymal Transition and Metastasis. Int J Radiat Oncol Biol Phys 2023; 115:719-732. [PMID: 36245124 DOI: 10.1016/j.ijrobp.2022.09.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE This study suggested that lenvatinib may incapacitate hepatocellular carcinoma (HCC) to radiation treatment by abrogating radiation-induced Src/signal transducer and the activator of transcription 3 signaling (STAT3)/nuclear factor-κB (NF-κB) to escalate radiation-induced extrinsic and intrinsic apoptosis. These findings uncover the role of targeting Src and its arbitrating epithelial-mesenchymal transition (EMT), which could increase the anti-HCC efficacy of radiation therapy (RT). Lenvatinib and sorafenib are multikinase inhibitors used to treat HCC. Lenvatinib is noninferior to sorafenib in the therapeutic response in HCC. However, whether lenvatinib intensifies the anti-HCC efficacy of RT is ambiguous. Several oncogenic kinases and transcription factors, such as Src, STAT3, and NF-κB, enhance the radiosensitivity of cancers. Therefore, we aimed to investigate the roles of the Src/STAT3/NF-κB axis in HCC after RT treatment and assessed whether targeting Src by lenvatinib may enhance the effectiveness of RT. METHODS AND MATERIALS Hep3B, Huh7, HepG2, and SK-Hep1 HCC cells and 2 types of animal models were used to identify the efficacy of RT combined with lenvatinib. Cellular toxicity, apoptosis, DNA damage, EMT/metastasis regulation, and treatment efficacy were validated by colony formation, flow cytometry, Western blotting, and in vivo experiments, respectively. Knockdown of Src by siRNA was also used to validate the role of Src in RT treatment. RESULTS Silencing Src reduced STAT3/NF-κB signaling and sensitized HCC to radiation. Lenvatinib reversed radiation-elicited Src/STAT3/NF-κB signaling while enhancing the anti-HCC efficacy of radiation. Both lenvatinib and siSrc promoted the radiation effect of cell proliferation on suppression, inhibition of the invasion ability, and induction of apoptosis in HCC. Lenvatinib also alleviated radiation-triggered oncogenic and EMT-related protein expression. CONCLUSIONS Our findings uncovered the role of the Src/STAT3/NF-κB regulatory axis in response to radiation-induced toxicity and confirmed Src as the key regulatory molecule for radiosensitization of HCC evoked by lenvatinib.
Collapse
Affiliation(s)
- Yueh-Shan Weng
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan
| | - I-Tsang Chiang
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan; Department of Medical Imaging and Radiologic Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan; Medical administrative center, Show Chwan Memorial Hospital, Changhua 500, Taiwan, ROC
| | - Jai-Jen Tsai
- Division of Gastroenterology, Department of Medicine, Taipei Veterans General Hospital, Yuan-Shan/Su-Ao Branch, Yi-Lan 260, Taiwan; Department of Medicine/Medical Research and Education, Taipei Veterans General Hospital, Yuan-Shan/Su-Ao Branch, Yi-Lan 260, Taiwan; Department of Nursing, Cardinal Tien Junior College of Healthcare and Management, New Taipei City 231, Taiwan
| | - Yu-Chang Liu
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua 500, Taiwan; Department of Medical Imaging and Radiologic Sciences, Central Taiwan University of Science and Technology, Taichung 406, Taiwan; Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Lukang, Changhua 505, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung 406, Taiwan.
| |
Collapse
|
3
|
van de Haterd B, Verboven K, Vandenabeele F, Agten A. The Role of Skeletal Muscle Mitochondria in Colorectal Cancer Related Cachexia: Friends or Foes? Int J Mol Sci 2022; 23:14833. [PMID: 36499157 PMCID: PMC9737299 DOI: 10.3390/ijms232314833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Up to 60% of colorectal cancer (CRC) patients develop cachexia. The presence of CRC related cachexia is associated with more adverse events during systemic therapy, leading to a high mortality rate. The main manifestation in CRC related cachexia is the loss of skeletal muscle mass, resulting from an imbalance between skeletal muscle protein synthesis and protein degradation. In CRC related cachexia, systemic inflammation, oxidative stress, and proteolytic systems lead to mitochondrial dysfunction, resulting in an imbalanced skeletal muscle metabolism. Mitochondria fulfill an important function in muscle maintenance. Thus, preservation of the skeletal muscle mitochondrial homeostasis may contribute to prevent the loss of muscle mass. However, it remains elusive whether mitochondria play a benign or malignant role in the development of cancer cachexia. This review summarizes current (mostly preclinical) evidence about the role of skeletal muscle mitochondria in the development of CRC related cachexia. Future human research is necessary to determine the physiological role of skeletal muscle mitochondria in the development of human CRC related cachexia.
Collapse
Affiliation(s)
- Britt van de Haterd
- REVAL—Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
| | - Kenneth Verboven
- REVAL—Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
- BIOMED—Biomedical Research Center, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
| | - Frank Vandenabeele
- REVAL—Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
| | - Anouk Agten
- REVAL—Rehabilitation Research Center, Faculty of Rehabilitation Sciences, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
- U-RISE—Uhasselt Research Group on Innovative and Society-Engaged Education, School for Educational Studies, Hasselt University, Agoralaan, 3590 Diepenbeek, Belgium
| |
Collapse
|
4
|
Piletska E, Magumba K, Joseph L, Garcia Cruz A, Norman R, Singh R, Tabasso AFS, Jones DJL, Macip S, Piletsky S. Molecular imprinting as a tool for determining molecular markers: a lung cancer case. RSC Adv 2022; 12:17747-17754. [PMID: 35765329 PMCID: PMC9200412 DOI: 10.1039/d2ra01830f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/23/2022] [Indexed: 11/29/2022] Open
Abstract
Determining which cancer patients will be sensitive to a given therapy is essential for personalised medicine. Thus, it is important to develop new tools that will allow us to stratify patients according to their predicted response to treatment. The aim of work presented here was to use molecular imprinting for determining the sensitivity of lung cancer cell lines to ionising radiation based on cell surface proteomic differences. Molecularly imprinted polymer nanoparticles (nanoMIPs) were formed in the presence of whole cells. Following trypsinolysis, protein epitopes protected by complexing with MIPs were eluted from the nanoparticles and analysed by LC-MS/MS. The analysis identified two membrane proteins, neutral amino acid transporter B (0) and 4F2 cell-surface antigen heavy chain, the abundance of which in the lung cancer cells could indicate resistance of these cells to radiotherapy. This proof-of-principle experiments shows that this technology can be used in the discovery of new biomarkers and in development of novel diagnostic and therapeutic tools for a personalised medicine approach to treating cancer. A first use of molecular imprinting for characterisation of surfaceome of the lung cancer cells and discovery of the molecular markers for radiosensitivity: towards development of an effective tool for cancer therapy and personalised medicine.![]()
Collapse
Affiliation(s)
- Elena Piletska
- School of Chemistry, College of Science and Engineering, University of Leicester Leicester UK +44-(0)116-0294-4666
| | - Kirabo Magumba
- School of Chemistry, College of Science and Engineering, University of Leicester Leicester UK +44-(0)116-0294-4666
| | - Lesslly Joseph
- School of Chemistry, College of Science and Engineering, University of Leicester Leicester UK +44-(0)116-0294-4666
| | - Alvaro Garcia Cruz
- School of Chemistry, College of Science and Engineering, University of Leicester Leicester UK +44-(0)116-0294-4666
| | - Rachel Norman
- Leicester Cancer Research Centre, University of Leicester Leicester Royal Infirmary Leicester UK
| | - Rajinder Singh
- Leicester Cancer Research Centre, University of Leicester Leicester Royal Infirmary Leicester UK
| | - Antonella F S Tabasso
- Leicester Cancer Research Centre, University of Leicester Leicester Royal Infirmary Leicester UK.,Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester UK
| | - Donald J L Jones
- Leicester Cancer Research Centre, University of Leicester Leicester Royal Infirmary Leicester UK.,Department of Cardiovascular Sciences, University of Leicester Leicester UK.,National Institute for Health Research, Leicester Biomedical Research Centre, Glenfield Hospital Leicester UK
| | - Salvador Macip
- Mechanisms of Cancer and Ageing Laboratory, Department of Molecular and Cell Biology, University of Leicester Leicester UK.,FoodLab, Faculty of Health Sciences, Universitat Oberta de Catalunya Barcelona Spain
| | - Sergey Piletsky
- School of Chemistry, College of Science and Engineering, University of Leicester Leicester UK +44-(0)116-0294-4666
| |
Collapse
|
5
|
The Impact of Weight Loss during Chemoradiotherapy for Unresectable Esophageal Cancer: Real-World Results. Life (Basel) 2022; 12:life12050706. [PMID: 35629373 PMCID: PMC9146706 DOI: 10.3390/life12050706] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022] Open
Abstract
Weight loss is a common phenomenon presented in unresectable esophageal cancer (EC) patients during their definitive chemoradiotherapy (dCRT) treatment course. This study explored the prognostic value of weight changes during dCRT in unresectable EC patients. From 2009 to 2017, 69 cT4b thoracic EC patients undergoing complete curative dCRT without baseline malnutrition were included. Clinical factors were analyzed via the Cox proportional hazards model and survival was analyzed by the Kaplan−Meier method. During dCRT, the median weight loss percentage was 5.51% (IQR = 2.77−8.85%), and the lowest body weight was reached at 35 days (IQR = 23−43 days). Median OS of these patients was 13.5 months. Both univariate and multivariate analysis demonstrated that weight loss ≤ 4% during dCRT was significantly associated with superior OS with a hazard ratio of 2.61 (95% CI: 1.40−4.85, p = 0.002). The median OS for patients with weight loss ≤ 4% and >4% during dCRT was 59.6 months and 9.7 months, respectively (p = 0.001). Our study demonstrated that weight loss ≤ 4% during dCRT course is a favorable prognostic factor for cT4b EC patients. This index could serve as a nutrition support reference for unresectable EC patients receiving dCRT in the future.
Collapse
|
6
|
Freire PP, Cury SS, Lopes LO, Fernandez GJ, Liu J, de Moraes LN, de Oliveira G, Oliveira JS, de Moraes D, Cabral-Marques O, Dal-Pai-Silva M, Hu X, Wang DZ, Carvalho RF. Decreased miR-497-5p Suppresses IL-6 Induced Atrophy in Muscle Cells. Cells 2021; 10:3527. [PMID: 34944037 PMCID: PMC8700610 DOI: 10.3390/cells10123527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022] Open
Abstract
Interleukin-6 (IL-6) is a pro-inflammatory cytokine associated with skeletal muscle wasting in cancer cachexia. The control of gene expression by microRNAs (miRNAs) in muscle wasting involves the regulation of thousands of target transcripts. However, the miRNA-target networks associated with IL6-induced muscle atrophy remain to be characterized. Here, we show that IL-6 promotes the atrophy of C2C12 myotubes and changes the expression of 20 miRNAs (5 up-regulated and 15 down-regulated). Gene Ontology analysis of predicted miRNAs targets revealed post-transcriptional regulation of genes involved in cell differentiation, apoptosis, migration, and catabolic processes. Next, we performed a meta-analysis of miRNA-published data that identified miR-497-5p, a down-regulated miRNAs induced by IL-6, also down-regulated in other muscle-wasting conditions. We used miR-497-5p mimics and inhibitors to explore the function of miR-497-5p in C2C12 myoblasts and myotubes. We found that miR-497-5p can regulate the expression of the cell cycle genes CcnD2 and CcnE1 without affecting the rate of myoblast cellular proliferation. Notably, miR-497-5p mimics induced myotube atrophy and reduced Insr expression. Treatment with miR-497-5p inhibitors did not change the diameter of the myotubes but increased the expression of its target genes Insr and Igf1r. These genes are known to regulate skeletal muscle regeneration and hypertrophy via insulin-like growth factor pathway and were up-regulated in cachectic muscle samples. Our miRNA-regulated network analysis revealed a potential role for miR-497-5p during IL6-induced muscle cell atrophy and suggests that miR-497-5p is likely involved in a compensatory mechanism of muscle atrophy in response to IL-6.
Collapse
Affiliation(s)
- Paula P. Freire
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
| | - Sarah S. Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Letícia O. Lopes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Geysson J. Fernandez
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
- Faculty of Medicine, University of Antioquia, UdeA, Medellín 050010, Colombia
| | - Jianming Liu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
| | - Leonardo Nazario de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Grasieli de Oliveira
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Jakeline S. Oliveira
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Diogo de Moraes
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Otavio Cabral-Marques
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil;
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, Brazil
- Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo 05508-000, Brazil
| | - Maeli Dal-Pai-Silva
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (J.L.); (X.H.); (D.-Z.W.)
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Robson F. Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University, UNESP, Botucatu 18618-689, Brazil; (P.P.F.); (S.S.C.); (L.O.L.); (G.J.F.); (L.N.d.M.); (G.d.O.); (J.S.O.); (D.d.M.); (M.D.-P.-S.)
| |
Collapse
|
7
|
Lokeshwar SD, Press BH, Nie J, Klaassen Z, Kenney PA, Leapman MS. Cachexia and bladder cancer: clinical impact and management. Curr Opin Support Palliat Care 2021; 15:260-265. [PMID: 34698663 DOI: 10.1097/spc.0000000000000580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to describe the causes, management, and clinical outcomes associated with cachexia and related components including sarcopenia, among patients with bladder cancer (BCa). RECENT FINDINGS Cachexia in patients with BCa is associated with poorer outcomes after radical cystectomy (RC), radiation, and chemotherapy. Nutritional supplements and novel pharmaceutical agents including magnolol, flucoidan and Anamorelin are currently undergoing investigation for their potential use in BCa patients with cachexia. SUMMARY Cachexia is a hypercatabolic state thought to be caused by an immune-regulated release of cytokines and disruptions of molecular pathways within the tumor microenvironment and systemically. Nutritional deficiencies in patients with BCa also contribute to cachexia and sarcopenia. Patients with BCa -related cachexia and sarcopenia experience worse survival and therapeutic outcomes after RC, chemotherapy, and radiation therapy. Patients with cachexia also experience more postoperative complications after RC. The management of cachexia in patients with BCa remains challenging and requires timely identification, and multidisciplinary management including nutritional supplementation, physical therapy, palliative care, and pharmacological agents. Clinical trials and human studies are still required to determine which pharmacological agents are optimal for BCa cachexia.
Collapse
Affiliation(s)
- Soum D Lokeshwar
- Department of Urology, Yale University School of Medicine, New Haven, Connecticut
| | - Benjamin H Press
- Department of Urology, Yale University School of Medicine, New Haven, Connecticut
| | - James Nie
- Department of Urology, Yale University School of Medicine, New Haven, Connecticut
| | - Zachary Klaassen
- Department of Urology, Medical College of Georgia, Augusta, Georgia, USA
| | - Patrick A Kenney
- Department of Urology, Yale University School of Medicine, New Haven, Connecticut
| | - Michael S Leapman
- Department of Urology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
8
|
L-carnitine ameliorates the muscle wasting of cancer cachexia through the AKT/FOXO3a/MaFbx axis. Nutr Metab (Lond) 2021; 18:98. [PMID: 34724970 PMCID: PMC8559414 DOI: 10.1186/s12986-021-00623-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/04/2021] [Indexed: 12/16/2022] Open
Abstract
Background Recent studies suggest potential benefits of applying L-carnitine in the treatment of cancer cachexia, but the precise mechanisms underlying these benefits remain unknown. This study was conducted to determine the mechanism by which L-carnitine reduces cancer cachexia. Methods C2C12 cells were differentiated into myotubes by growing them in DMEM for 24 h (hrs) and then changing the media to DMEM supplemented with 2% horse serum. Differentiated myotubes were treated for 2 h with TNF-α to establish a muscle atrophy cell model. After treated with L-carnitine, protein expression of MuRF1, MaFbx, FOXO3, p-FOXO3a, Akt, p-Akt, p70S6K and p-p70S6K was determined by Western blotting. Then siRNA-Akt was used to determine that L-carnitine ameliorated cancer cachexia via the Akt/FOXO3/MaFbx. In vivo, the cancer cachexia model was established by subcutaneously transplanting CT26 cells into the left flanks of the BALB/c nude mice. After treated with L-carnitine, serum levels of IL-1, IL-6 and TNF-α, and the skeletal muscle content of MuRF1, MaFbx, FOXO3, p-FOXO3a, Akt, p-Akt, p70S6K and p-p70S6K were measured. Results L-carnitine increased the gastrocnemius muscle (GM) weight in the CT26-bearing cachexia mouse model and the cross-sectional fiber area of the GM and myotube diameters of C2C12 cells treated with TNF-α. Additionally, L-carnitine reduced the protein expression of MuRF1, MaFbx and FOXO3a, and increased the p-FOXO3a level in vivo and in vitro. Inhibition of Akt, upstream of FOXO3a, reversed the effects of L-carnitine on the FOXO3a/MaFbx pathway and myotube diameters, without affecting FOXO3a/MuRF-1. In addition to regulating the ubiquitination of muscle proteins, L-carnitine also increased the levels of p-p70S6K and p70S6K, which are involved in protein synthesis. Akt inhibition did not reverse the effects of L-carnitine on p70S6K and p-p70S6K. Hence, L-carnitine ameliorated cancer cachexia via the Akt/FOXO3/MaFbx and p70S6K pathways. Moreover, L-carnitine reduced the serum levels of IL-1 and IL-6, factors known to induce cancer cachexia. However, there were minimal effects on TNF-α, another inducer of cachexia, in the in vivo model. Conclusion These results revealed a novel mechanism by which L-carnitine protects muscle cells and reduces inflammation related to cancer cachexia.
Collapse
|
9
|
Chen MF, Hsieh CC, Chen PT, Lu MS. Role of Nutritional Status in the Treatment Outcome for Esophageal Squamous Cell Carcinoma. Nutrients 2021; 13:2997. [PMID: 34578883 PMCID: PMC8466664 DOI: 10.3390/nu13092997] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Undernourishment is reported to impair treatment response, further leading to poor prognosis for cancer patients. We aimed to investigate the role of nutritional status on the prognosis of squamous cell carcinoma (SCC) of the esophagus, and its correlation with anticancer immune responsiveness. We retrospectively reviewed 340 esophageal-SCC patients who completed curative treatment and received a nutrition evaluation by the Patient-Generated Subjective Global Assessment (PGSGA) score at the beginning and completion of neoadjuvant treatment at our hospital. The correlation between the nutritional status and various clinicopathological parameters and prognosis were examined. In addition, the role of nutritional status in the regulation of the anticancer immune response was also assessed in cancer patients and in a 4-nitroquinoline 1-oxide (4NQO)-induced esophageal tumor model. Our data revealed that malnutrition (patients with a high PGSGA score) was associated with advanced stage and reduced survival rate. Patients in the group with a high PGSGA score were correlated with the higher neutrophil-to-lymphocyte ratio, higher proportion of myeloid-derived-suppressor cells (MDSC) and increased IL-6 level. Furthermore, surgical resection brought the survival benefit to patients in the low PGSGA group, but not for the malnourished patients after neoadjuvant treatment. Using a 4NQO-induced tumor model, we found that nutrition supplementation decreased the rate of invasive tumor formation and attenuated the immune-suppressive microenvironment. In conclusion, malnutrition was associated with poor prognosis in esophageal-SCC patients. Nutritional status evaluated by PGSGA may be useful to guide treatment decisions in clinical practice. Nutritional supplementation is suggested to improve prognosis, and it might be related to augmented anticancer immune response.
Collapse
Affiliation(s)
- Miao-Fen Chen
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Ching-Chuan Hsieh
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Ping-Tsung Chen
- Department of Medical Oncology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
| | - Ming-Shian Lu
- Department of Thoracic & Cardiovascular Surgery, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan;
| |
Collapse
|
10
|
Kowshik V, Velkumary S, Sethi P, Feula JM, Subhashri S, Abirami M. Association of handgrip strength and endurance with body composition in head and neck cancer patients. J Family Med Prim Care 2021; 10:910-916. [PMID: 34041097 PMCID: PMC8138385 DOI: 10.4103/jfmpc.jfmpc_1695_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/27/2020] [Accepted: 12/16/2020] [Indexed: 01/10/2023] Open
Abstract
Introduction: Assessment of skeletal muscle function (SMF) is of clinical relevance in the prediction of treatment outcome and to decide on optimal management of head & neck cancer (HNC) patients. Handgrip strength (HGS) & handgrip endurance (HGE) are considered as surrogate marker for whole-body skeletal muscle function. Further, SMF depends substantially on the body composition (BC). Hence in this study, we compared BC, HGS and HGE between HNC patients and healthy controls and also analysed the association of HGS, HGE with body composition in HNC patients. Methods: A cross-sectional study, conducted in 44 subjects in the age between 18 to 60 years. Twenty-two were histologically proven HNC patients prior to cancer-specific treatment and twenty-two age and gender-matched healthy volunteers. The parameters recorded were Height, weight, waist circumference, hip circumference, HGS, HGE and BC. Hand-held dynamometer was used to measure HGS and HGE measured using a stopwatch. BC was estimated by whole-body bioelectrical Impedance analysis method using Bodystat Quad scan 4000 device. Result: Comparison of data between HNC patient & healthy control was done by Student's t test. HGS, HGE, lean body mass (LBM), fat-free mass index (FFMI), Phase angle (PA), body cell mass (BCM) and body cell mass index (BCMI) were found to be reduced significantly in HNC patients when compared to healthy subjects. Further, Pearson correlation analysis revealed a significant positive correlation of HGS & HGE with LBM, FFMI, PA, BCM & BCMI, whereas body fat mass index showed a negative correlation with HGS & HGE in HNC patients. Conclusion: Our findings revealed, a significant reduction in HGS, HGE in patient with HNC which denotes decreased skeletal muscle function and it is linearly associated with low muscle mass, body cell mass and phase angle.
Collapse
Affiliation(s)
- Vengadesan Kowshik
- 3rd Year MBBS, Department of Physiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Subramanian Velkumary
- Additional Professor, Department of Physiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Pooja Sethi
- Assistant Professor, Department of Radiation Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Jothi Marie Feula
- Assitant Professor, Department of Physiology, Aarupadai Veedu Medical College and Hospital, Puducherry, India
| | - Soundirarajan Subhashri
- Assistant Professor, Department of Physiology, Sri Venkateshwaraa Medical College Hospital and Research Centre, Puducherry, India
| | - Manikandan Abirami
- III Year Post Graduate, Department of Physiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| |
Collapse
|
11
|
Baliga MS, Rao S, Hegde SK, Rao P, Simon P, George T, Venkatesh P, Baliga-Rao MP, Thilakchand KR. Usefulness of Honey as an Adjunct in the Radiation Treatment for Head and Neck Cancer: Emphasis on Pharmacological and Mechanism/s of Actions. Anticancer Agents Med Chem 2021; 22:20-29. [PMID: 33573581 DOI: 10.2174/1871520621666210126094509] [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] [Received: 04/30/2020] [Revised: 07/13/2020] [Accepted: 07/19/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In the treatment of head and neck cancer (HNC), ionizing radiation is an important modality in achieving curative objectives. However, the effective use of radiation is compromised by the side effects resulting from the damage to the adjacent normal tissue. Preclinical studies carried out in the recent past have shown that the age old dietary agent honey, which also possess myriad medicinal use is beneficial in mitigating diverse radiation-induced side effects like mucositis, xerostomia, fatigue, weight loss and to promote healing of refractory wounds. OBJECTIVE The objective of this memoir is to review the beneficial effects of honey in mitigating radiation-induced side effects in HNC and to emphasize on the underlying mechanism of action for the beneficial effects Methods: Two authors searched Google Scholar, PubMed, Embase, and the Cochrane Library for publications up to December 2019 to assess the ability of honey in reducing the severity of radiation-induced ill effects in the treatment of HNC. Subsequently, the adjunct pharmacological effects and mechanism/s responsible were also searched for and appropriately used to substantiate the underlying mechanism/s of action for the beneficial effects. RESULTS The existing data is suggestive that honey is beneficial in mitigating the radiation-induced mucositis, xerostomia, healing of recalcitrant wounds in radiation exposed regions and multiple pathways mediate the beneficial effects especially, free radical scavenging, antioxidant, wound healing, anticancer, analgesic, anti-inflammatory, anabolic, anti-fatigue and anti-anaemic effects that add additional value to the use of honey as an adjunct in cancer therapy. CONCLUSION For the first time this review addresses the underlying pharmacological effects related to the beneficial effects of honey in radiation-induced damage, and attempts at emphasizes the lacunae that need further studies for optimizing the use of honey as an adjunct in radiotherapy of HNC. The authors suggest that future studies should be directed at understanding the detail molecular mechanisms responsible for the beneficial effects using validated cell culture and animal models of study. Large multi centric clinical trials with standardised honey is also needed to understand the clinical use of honey.
Collapse
Affiliation(s)
- Manjeshwar S Baliga
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Suresh Rao
- Radiation Oncology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Sanath K Hegde
- Radiation Oncology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Pratima Rao
- Department of Orodental Pathology, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Paul Simon
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Thomas George
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | - Ponemone Venkatesh
- Research Unit, Mangalore Institute of Oncology, Pumpwell, Mangalore, Karnataka. India
| | | | - Karadka R Thilakchand
- Department of Anesthesiology, Karnataka Institute of Medical Sciences, Hubballi 580022. India
| |
Collapse
|
12
|
Lim S, Brown JL, Washington TA, Greene NP. Development and progression of cancer cachexia: Perspectives from bench to bedside. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:177-185. [PMID: 34447946 PMCID: PMC8386816 DOI: 10.1016/j.smhs.2020.10.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/24/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cachexia (CC) is a devastating syndrome characterized by weight loss, reduced fat mass and muscle mass that affects approximately 80% of cancer patients and is responsible for 22%-30% of cancer-associated deaths. Understanding underlying mechanisms for the development of CC are crucial to advance therapies to treat CC and improve cancer outcomes. CC is a multi-organ syndrome that results in extensive skeletal muscle and adipose tissue wasting; however, CC can impair other organs such as the liver, heart, brain, and bone as well. A considerable amount of CC research focuses on changes that occur within the muscle, but cancer-related impairments in other organ systems are understudied. Furthermore, metabolic changes in organ systems other than muscle may contribute to CC. Therefore, the purpose of this review is to address degenerative mechanisms which occur during CC from a whole-body perspective. Outlining the information known about metabolic changes that occur in response to cancer is necessary to develop and enhance therapies to treat CC. As much of the current evidences in CC are from pre-clinical models we should note the majority of the data reviewed here are from preclinical models.
Collapse
Affiliation(s)
- Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, 155 Stadium Dr, Fayetteville, AR, USA
| | - Jacob L. Brown
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK, USA
| | - Tyrone A. Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, 155 Stadium Dr, Fayetteville, AR, USA
| | - Nicholas P. Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, 155 Stadium Dr, Fayetteville, AR, USA
| |
Collapse
|
13
|
Polymorphism of TNFRSF1 A may act as a predictor of severe radiation-induced oral mucositis and a prognosis factor in patients with head and neck cancer. Oral Surg Oral Med Oral Pathol Oral Radiol 2020; 130:283-291.e2. [PMID: 32561252 DOI: 10.1016/j.oooo.2020.05.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the relationship between single nucleotide polymorphism (SNP) (-135 T>C) of TNFRSF1 A and the frequency of occurrence and severity of oral mucositis (OM) in patients with head and neck cancer (HNC) treated with radiotherapy (RT). STUDY DESIGN This retrospective, cohort study included 60 patients with HNC treated with intensity-modulated radiation therapy (IMRT). TNFRSF1 A SNP analysis (-135 T>C) was performed by using molecular probes (TaqMan, ThermoFisher Scientific, Waltham, MA) in DNA isolated from peripheral blood (QIAamp DNA MiniKit; Qiagen, Germantown, MD). RESULTS CC genotype was related to 4.5-fold higher risk of grade 2 OM after the second week of RT. Similarly, CC carriers had a significantly higher risk of severe (grade 3) OM after the fourth (6-fold) and fifth (7.5-fold) weeks of RT. The CC genotype of the TNFRSF1 A gene was significantly correlated with a higher risk of shorter overall survival (OS) (> 37 months follow-up period; hazard ratio [HR] = 2.78). CONCLUSIONS SNP (-135 T>C) of the TNFRSF1 A gene may act as a predictor of OM occurrence in patients with HNC treated with IMRT. The studied SNP may also serve as a prognostic factor in such cases.
Collapse
|
14
|
Sun A, Xu K, Liu H, Li H, Shi Y, Zhu X, Liang T, Li X, Cao X, Ji Y, Jiang T, Xu C, Liu X. The evolution of zebrafish RAG2 protein is required for adapting to the elevated body temperature of the higher endothermic vertebrates. Sci Rep 2020; 10:4126. [PMID: 32139788 PMCID: PMC7057966 DOI: 10.1038/s41598-020-61019-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/19/2020] [Indexed: 11/21/2022] Open
Abstract
The recombination activating gene (RAG or RAG1/RAG2 complex)-mediated adaptive immune system is a hallmark of jawed vertebrates. It has been reported that RAG originated in invertebrates. However, whether RAG further evolved once it arose in jawed vertebrates remains largely unknown. Here, we found that zebrafish RAG (zRAG) had a lower activity than mouse RAG (mRAG). Intriguingly, the attenuated stability of zebrafish RAG2 (zRAG2), but not zebrafish RAG1, caused the reduced V(D)J recombination efficiency compared to mRAG at 37 °C which are the body temperature of most endotherms except birds. Importantly, the lower temperature 28 °C, which is the best temperature for zebrafish growth, made the recombination efficiency of zRAG similar to that of mRAG by improving the stability of zRAG2. Consistent with the prementioned observation, the V(D)J recombination of Rag2KI/KI mice, which zRAG2 was substituted for mRAG2, was also severely impaired. Unexpectedly, Rag2KI/KI mice developed cachexia syndromes accompanied by premature death. Taken together, our findings illustrate that the evolution of zebrafish RAG2 protein is required for adapting to the elevated body temperature of the higher endothermic vertebrates.
Collapse
Affiliation(s)
- Ao Sun
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ke Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haifeng Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hua Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yaohuang Shi
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Xiaoyan Zhu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Tao Liang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xinyue Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xianxia Cao
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yanhong Ji
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Centre, Xi'an, Shaanxi, 710061, China
| | - Taijiao Jiang
- Center of System Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Chenqi Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaolong Liu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
15
|
Konings K, Vandevoorde C, Baselet B, Baatout S, Moreels M. Combination Therapy With Charged Particles and Molecular Targeting: A Promising Avenue to Overcome Radioresistance. Front Oncol 2020; 10:128. [PMID: 32117774 PMCID: PMC7033551 DOI: 10.3389/fonc.2020.00128] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/24/2020] [Indexed: 12/13/2022] Open
Abstract
Radiotherapy plays a central role in the treatment of cancer patients. Over the past decades, remarkable technological progress has been made in the field of conventional radiotherapy. In addition, the use of charged particles (e.g., protons and carbon ions) makes it possible to further improve dose deposition to the tumor, while sparing the surrounding healthy tissues. Despite these improvements, radioresistance and tumor recurrence are still observed. Although the mechanisms underlying resistance to conventional radiotherapy are well-studied, scientific evidence on the impact of charged particle therapy on cancer cell radioresistance is restricted. The purpose of this review is to discuss the potential role that charged particles could play to overcome radioresistance. This review will focus on hypoxia, cancer stem cells, and specific signaling pathways of EGFR, NFκB, and Hedgehog as well as DNA damage signaling involving PARP, as mechanisms of radioresistance for which pharmacological targets have been identified. Finally, new lines of future research will be proposed, with a focus on novel molecular inhibitors that could be used in combination with charged particle therapy as a novel treatment option for radioresistant tumors.
Collapse
Affiliation(s)
- Katrien Konings
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Department of Nuclear Medicine, iThemba LABS, Cape Town, South Africa
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| | - Sarah Baatout
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium.,Department of Molecular Biotechnology, Ghent University, Ghent, Belgium
| | - Marjan Moreels
- Radiobiology Unit, Belgian Nuclear Research Center (SCK•CEN), Mol, Belgium
| |
Collapse
|
16
|
Astradsson T, Sellberg F, Berglund D, Ehrsson YT, Laurell GFE. Systemic Inflammatory Reaction in Patients With Head and Neck Cancer-An Explorative Study. Front Oncol 2019; 9:1177. [PMID: 31750257 PMCID: PMC6848384 DOI: 10.3389/fonc.2019.01177] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
Aim: To assess the longitudinal pattern of pro-inflammatory cytokines and growth factors in serum up to 1 year following treatment for head and neck cancer. Materials and Methods: Patients with newly diagnosed, curable head and neck cancer were included (n = 30). The most common subsite was oropharynx (n = 13) followed by oral cavity (n = 9). Blood was drawn from all patients at regular intervals (before treatment, 7 weeks after the start of the treatment, and at 3 months and 1 year after termination of treatment) and analyzed for cytokines (Il-1β, Il-2, Il-4, Il-5, Il-6, Il-8, Il-10, GM-CSF, TNF-α, and IFN-γ) and growth factors (G-CSF, FGF-2, EGF, and VEGF). Results: The time point of the peak level of pro-inflammatory cytokines was 7 weeks after start of treatment which corresponded for the majority of patients with termination of radiotherapy or chemoradiotherapy. Patients undergoing chemoradiotherapy exhibited a significant increase of IL-1β, IL-6, and IL-10 at 7 weeks as compared to pre-treatment levels. At 1 year after termination of treatment four patients experienced recurrence of disease while 26 patients were considered disease-free. The patients with recurrence had significantly higher levels of IL-1β, IL-6, IL-8, and IL-10 at 7 weeks after the start of treatment than patients without recurrence. Correlated with T stadium patients with T3-T4 had higher levels of IL-1β and IL-8 than patients with T1-T2 7 weeks after the start of treatment. Conclusions: The observed immune response in this explorative study demonstrates that chemoradiotherapy may induce not only a local treatment effect on the immune system but also effects far outside the irradiated field. The result of the study indicates that analysis of a pro-inflammatory panel of cytokines in serum at 7 weeks after the start of treatment could be of prognostic value in patients with head and neck cancer. Further study of a larger cohort could help identify patients at larger risk for recurrent disease with measurements of pro-inflammatory cytokines under and after treatment.
Collapse
Affiliation(s)
| | - Felix Sellberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - David Berglund
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | | |
Collapse
|
17
|
Abstract
Cancer is a catabolic inflammatory disease that causes patients to often experience weight loss, or even cachexia in severe cases. Undernourishment in patients with cancer impairs the quality of life and therapeutic response, further leading to poor prognosis. Active and frequent nutritional screening and assessment using valid tools are important for fast and appropriate nutritional intervention. Additionally, a suitable individualized nutritional intervention strategy should be established based on the nutritional assessment result. In general, nutritional intervention begins with nutritional counseling of patients diagnosed with cancer, and a well-planned nutritional counseling improves the treatment adherence and nutritional status. When planning nutritional supplementation for cancer patients, specific nutrients, including amino acids and fatty acids, should be considered. However, there has been no consistent result showing that any particular nutrient significantly improves the prognosis of cancer patients. Hence, continuous attention from clinical physicians is needed to plan nutritional improvement in patients with cancer.
Collapse
Affiliation(s)
- Duk Hwan Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| |
Collapse
|
18
|
Duong L, Radley-Crabb HG, Gardner JK, Tomay F, Dye DE, Grounds MD, Pixley FJ, Nelson DJ, Jackaman C. Macrophage Depletion in Elderly Mice Improves Response to Tumor Immunotherapy, Increases Anti-tumor T Cell Activity and Reduces Treatment-Induced Cachexia. Front Genet 2018; 9:526. [PMID: 30459812 PMCID: PMC6232269 DOI: 10.3389/fgene.2018.00526] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/19/2018] [Indexed: 01/06/2023] Open
Abstract
Most cancers emerge in the elderly, including lung cancer and mesothelioma, yet the elderly remain an underrepresented population in pre-clinical cancer studies and clinical trials. The immune system plays a critical role in the effectiveness of many anti-cancer therapies in young hosts via tumor-specific T cells. However, immunosuppressive macrophages can constitute up to 50% of the tumor burden and impair anti-tumor T cell activity. Altered macrophage phenotype and function during aging may further impact anti-tumor T cell responses. Yet, the impact of macrophages on anti-tumor T cell responses and immunotherapy in the elderly is unknown. Therefore, we examined macrophages and their interaction with T cells in young (3 months) and elderly (20-24 months) AE17 mesothelioma-bearing female C57BL/6J mice during tumor growth. Mesothelioma tumors grew faster in elderly compared with young mice, and this corresponded with an increase in tumor-associated macrophages. During healthy aging, macrophages increase in bone marrow and spleens suggesting that these sites have an increased potential to supply cancer-promoting macrophages. Interestingly, in tumor-bearing mice, bone marrow macrophages increased proliferation whilst splenic macrophages had reduced proliferation in elderly compared with young mice, and macrophage depletion using the F4/80 antibody slowed tumor growth in young and elderly mice. We also examined responses to treatment with intra-tumoral IL-2/anti-CD40 antibody immunotherapy and found it was less effective in elderly (38% tumor regression) compared to young mice (90% regression). Tumor-bearing elderly mice decreased in vivo anti-tumor cytotoxic T cell activity in tumor draining lymph nodes and spleens. Depletion of macrophages using F4/80 antibody in elderly, but not young mice, improved IL-2/anti-CD40 immunotherapy up to 78% tumor regression. Macrophage depletion also increased in vivo anti-tumor T cell activity in elderly, but not young mice. All the tumor-bearing elderly (but not young) mice had decreased body weight (i.e., exhibited cachexia), which was greatly exacerbated by immunotherapy; whereas macrophage depletion prevented this immunotherapy-induced cachexia. These studies strongly indicate that age-related changes in macrophages play a key role in driving cancer cachexia in the elderly, particularly during immunotherapy, and sabotage elderly anti-tumor immune responses.
Collapse
Affiliation(s)
- Lelinh Duong
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Hannah G Radley-Crabb
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Joanne K Gardner
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Federica Tomay
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Danielle E Dye
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Miranda D Grounds
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Fiona J Pixley
- School of Biomedical Sciences, the University of Western Australia, Perth, WA, Australia
| | - Delia J Nelson
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Connie Jackaman
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| |
Collapse
|
19
|
Myokines as Possible Therapeutic Targets in Cancer Cachexia. J Immunol Res 2018; 2018:8260742. [PMID: 30426026 PMCID: PMC6217752 DOI: 10.1155/2018/8260742] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 09/23/2018] [Indexed: 01/04/2023] Open
Abstract
Cachexia is an extremely serious syndrome which occurs in most patients with different cancers, and it is characterized by systemic inflammation, a negative protein and energy balance, and involuntary loss of body mass. This syndrome has a dramatic impact on the patient's quality of life, and it is also associated with a low response to chemotherapy leading to a decrease in survival. Despite this, cachexia is still underestimated and often untreated. New research is needed in this area to understand this complex phenomenon and ultimately find treatment methods and therapeutic targets. The skeletal muscle can act as an endocrine organ. Signaling between muscles and other systems is done through myokines, cytokines, and proteins produced and released by myocytes. In this review, we would like to draw attention to some of the most important myokines that could have potential as biomarkers and therapeutic targets: myostatin, irisin, myonectin, decorin, fibroblast growth factor 21, interleukin-6, interleukin-8, and interleukin-15.
Collapse
|
20
|
Oei RW, Ye L, Kong F, Du C, Zhai R, Xu T, Shen C, Wang X, He X, Kong L, Hu C, Ying H. Prognostic value of inflammation-based prognostic index in patients with nasopharyngeal carcinoma: a propensity score matching study. Cancer Manag Res 2018; 10:2785-2797. [PMID: 30147375 PMCID: PMC6103307 DOI: 10.2147/cmar.s171239] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose The aim of this article is to investigate the significance of pretreatment prognostic nutritional index (PNI), systemic immune-inflammation index (SII), and their combination in nasopharyngeal carcinoma (NPC) patients receiving intensity-modulated radiotherapy (IMRT). Materials and methods A total of 585 patients were included. PNI and SII were calculated within 2 weeks prior to treatment. The optimal cutoff points were determined based on receiver operating characteristics curve analysis. The correlation between variables was analyzed. Kaplan–Meier method and Cox proportional hazards model were performed to evaluate the impact of both indices on overall survival (OS), progression-free survival (PFS) and distant metastasis-free survival (DMFS). Further propensity score matching (PSM) was carried out to minimize the effects of confounders. Results The optimal cutoff point of 53.0 for PNI and 527.20 for SII were selected. Pearson correlation coefficient showed an inverse correlation between PNI and SII (r = −0.232, P < 0.001). Multivariate analysis demonstrated that pretreatment PNI was an independent prognostic factor for OS (P = 0.047) and DMFS (P = 0.002) while pretreatment SII was an independent prognostic factor for OS (P = 0.003), PFS (P = 0.002), and DMFS (P = 0.002). After PSM, both parameters remained as independent prognosticators of survival. Additional prognostic value was observed in the combined use of PNI and SII. Conclusion Pretreatment PNI and SII are promising indicators of survival in NPC patients undergoing IMRT. They can be utilized to refine current TNM staging system in predicting prognosis and developing an individualized treatment in these patients.
Collapse
Affiliation(s)
- Ronald Wihal Oei
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Lulu Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Fangfang Kong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Chengrun Du
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Ruiping Zhai
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Tingting Xu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Chunying Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Xiaoshen Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Xiayun He
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Lin Kong
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Chaosu Hu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| | - Hongmei Ying
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, People's Republic of China, .,Department of Oncology, Shanghai Medical College, Shanghai, People's Republic of China,
| |
Collapse
|
21
|
de Sire R, Rizzatti G, Ingravalle F, Pizzoferrato M, Petito V, Lopetuso L, Graziani C, de Sire A, Mentella MC, Mele MC, Gasbarrini A, Scaldaferri F. Skeletal muscle-gut axis: emerging mechanisms of sarcopenia for intestinal and extra intestinal diseases. MINERVA GASTROENTERO 2018; 64:351-362. [PMID: 30016852 DOI: 10.23736/s1121-421x.18.02511-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In recent years, there has been an increasing interest on muscle wasting, considering the reduction of quality of life and the increase of morbidity and mortality associated. Sarcopenia and cachexia represent two conditions of reduction of muscle mass, sharing several elements involved in their pathogenesis, such as systemic inflammation, impaired muscle protein synthesis, increased muscle apoptosis, mitochondrial dysfunction in skeletal muscle tissue and insulin resistance. These features often characterize cancer, inactivity or denervation, but also inflammatory diseases, such as chronic obstructive pulmonary disease, renal failure, cardiac failure, rheumatoid arthritis, inflammatory bowel disease and aging in general. The gastrointestinal tract and gut microbiota are thought to be deeply associated with muscle function and metabolism, although the exact mechanisms that link gut with skeletal muscle are still not well known. This review summarized the potential pathways linking gut with muscle, in particular in conditions as sarcopenia and cachexia. The main emerging pathways implicated in the skeletal muscle-gut axis are: the myostatin/activin signaling pathway, the IGF1/PI3K/AKT/mTOR signaling pathway, which results suppressed, the NF-kB signaling pathway and the FOXO signaling pathway. Further researches in this field are necessary to better explain the linkage between gut microbiota and muscle wasting and the possible emerging therapies associated.
Collapse
Affiliation(s)
- Roberto de Sire
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy
| | - Gianenrico Rizzatti
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy
| | - Fabio Ingravalle
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy
| | - Marco Pizzoferrato
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy.,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Petito
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy.,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Loris Lopetuso
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy.,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristina Graziani
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy.,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alessandro de Sire
- Department of Medical and Surgical Specialties and Dentistry, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Chiara Mentella
- Clinical Nutrition, A. Gemelli Polyclinic Foundation and Institute for Research and Care, Rome, Italy
| | - Maria Cristina Mele
- Clinical Nutrition, A. Gemelli Polyclinic Foundation and Institute for Research and Care, Rome, Italy
| | - Antonio Gasbarrini
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy.,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Franco Scaldaferri
- Istituto di Patologia Speciale Medica, Sacred Heart Catholic University, Rome, Italy - .,UOC Medicina Interna e Gastroenterologia, Area Gastroenterologia e Oncologia Medica, Dipartimento di Scienze Gastroenterologiche, Endocrino-Metaboliche e Nefro-Urologiche, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW Cancer-associated weight loss is a common comorbid condition best described among patients with advanced malignancy receiving systemic therapy, but its relationship to patients undergoing radiation treatment is less well described. We review the interaction between cancer-associated weight loss and radiation treatment as well as its prognostic significance. RECENT FINDINGS Multiple studies demonstrate a consistent detrimental effect of cancer-associated weight loss either existing prior to treatment or developing during radiotherapy. Emerging data suggest cancer-associated weight loss independently contributes to an aggressive malignant phenotype rather than simply reflecting a consequence of disease. Novel therapies are urgently needed to address the unmet burden of cancer-associated weight loss. SUMMARY Consideration of cancer-associated weight loss is important among patients receiving radiotherapy. Further study will further characterize the relationship and identify targetable biologic mechanisms of cancer cachexia.
Collapse
|
23
|
Voltarelli FA, Frajacomo FT, Padilha CDS, Testa MTJ, Cella PS, Ribeiro DF, de Oliveira DX, Veronez LC, Bisson GS, Moura FA, Deminice R. Syngeneic B16F10 Melanoma Causes Cachexia and Impaired Skeletal Muscle Strength and Locomotor Activity in Mice. Front Physiol 2017; 8:715. [PMID: 29033844 PMCID: PMC5626871 DOI: 10.3389/fphys.2017.00715] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022] Open
Abstract
Muscle wasting has been emerging as one of the principal components of cancer cachexia, leading to progressive impairment of work capacity. Despite early stages melanomas rarely promotes weight loss, the appearance of metastatic and/or solid tumor melanoma can leads to cachexia development. Here, we investigated the B16F10 tumor-induced cachexia and its contribution to muscle strength and locomotor-like activity impairment. C57BL/6 mice were subcutaneously injected with 5 × 104 B16F10 melanoma cells or PBS as a Sham negative control. Tumor growth was monitored during a period of 28 days. Compared to Sham mice, tumor group depicts a loss of skeletal muscle, as well as significantly reduced muscle grip strength and epididymal fat mass. This data are in agreement with mild to severe catabolic host response promoted by elevated serum tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and lactate dehydrogenase (LDH) activity. Tumor implantation has also compromised general locomotor activity and decreased exploratory behavior. Likewise, muscle loss, and elevated inflammatory interleukin were associated to muscle strength loss and locomotor activity impairment. In conclusion, our data demonstrated that subcutaneous B16F10 melanoma tumor-driven catabolic state in response to a pro-inflammatory environment that is associated with impaired skeletal muscle strength and decreased locomotor activity in tumor-bearing mice.
Collapse
Affiliation(s)
- Fabrício A Voltarelli
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil.,Department of Physical Education, Faculty of Physical Education, Federal University of Mato GrossoCuiabá, Brazil
| | - Fernando T Frajacomo
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil.,Program of Molecular Carcinogenesis, Brazilian National Institute of CancerRio de Janeiro, Brazil
| | - Camila de Souza Padilha
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Mayra T J Testa
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Paola S Cella
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Diogo F Ribeiro
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Donizete X de Oliveira
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Luciana C Veronez
- Department of Maternal-Infant Nursing and Public Health, Ribeirao Preto College of Nursing, University of São PauloSão Paulo, Brazil
| | - Gabriela S Bisson
- Department of Maternal-Infant Nursing and Public Health, Ribeirao Preto College of Nursing, University of São PauloSão Paulo, Brazil
| | - Felipe A Moura
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| | - Rafael Deminice
- Department of Physical Education, Faculty of Physical Education and Sport, State University of LondrinaLondrina, Brazil
| |
Collapse
|
24
|
Pang Q, Jin H, Qu K, Man Z, Wang Y, Yang S, Zhou L, Liu H. The effects of nonsteroidal anti-inflammatory drugs in the incident and recurrent risk of hepatocellular carcinoma: a meta-analysis. Onco Targets Ther 2017; 10:4645-4656. [PMID: 29033583 PMCID: PMC5614766 DOI: 10.2147/ott.s143154] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Recent studies have showed that nonsteroidal anti-inflammatory drugs (NSAIDs) could reduce the risk of several types of cancer. However, epidemiological evidence of the association between NSAIDs intake and the risk of hepatocellular carcinoma (HCC) remains controversial. Methods To assess the preventive benefit of NSAIDs in HCC, we simultaneously searched the databases of PubMed, EmBase, Web of Science, and Scopus and screened eligible publications. Results A total of twelve articles (published from 2000 to 2017) from five countries were identified by retrieval. We observed a significantly lower risk of HCC incidence among users of NSAIDs than among those who did not use NSAIDs (pooled hazard ratio [HR] value =0.81, 95% confidence interval [CI]: 0.69–0.94). No evidence of publication bias was observed (Begg’s test, P=0.755; Egger’s test, P=0.564). However, when stratified according to the categories of NSAIDs, users of non-aspirin NSAIDs (HR =0.81, 95% CI: 0.70–0.94), but not aspirin (HR =0.77, 95% CI: 0.58–1.02), showed a statistically significant reduced HCC incidence. We also found that NSAIDs use significantly reduced the recurrent risk of HCC, with a HR value of 0.79 (95% CI: 0.75–0.84), whereas there was no statistically significant association between NSAIDs use and HCC mortality, with a HR value 0.65 (95% CI: 0.40–1.06). Conclusion Taken together, our meta-analysis demonstrates that NSAIDs significantly reduce the incident and recurrent risk of HCC.
Collapse
Affiliation(s)
- Qing Pang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Hao Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Kai Qu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhongran Man
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Yong Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Song Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Lei Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| | - Huichun Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui
| |
Collapse
|
25
|
The immunosuppressive cytokine interleukin-4 increases the clonogenic potential of prostate stem-like cells by activation of STAT6 signalling. Oncogenesis 2017; 6:e342. [PMID: 28553931 PMCID: PMC5523058 DOI: 10.1038/oncsis.2017.23] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/24/2017] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
Interleukin-4 plays a critical role in the regulation of immune responses and has been detected at high levels in the tumour microenvironment of cancer patients, where concentrations correlate with the grade of malignancy. In prostate cancer, interleukin-4 has been associated with activation of the androgen receptor, increased proliferation and activation of survival pathways such as Akt and NF-κB. However, its role in therapy resistance has not yet been determined. Here we investigate the influence of interleukin-4 on primary epithelial cells from prostate cancer patients. Our data demonstrate an increase in the clonogenic potential of these cells when cultured in the presence of interleukin-4. In addition, a Phospho-Kinase Array revealed that in contrast to previously published work, signal transducer and activator of transcription6 (STAT6) is the only signalling molecule activated after interleukin-4 treatment. Using the STAT6-specific inhibitor AS1517499 we could confirm the role of STAT6 in increasing colony-forming frequency. However, clonogenic recovery assays revealed that interleukin-4 does not rescue the effects of either irradiation or docetaxel treatment. We therefore propose that although the interleukin-4/STAT6 axis does not appear to be involved in therapy resistance, it does play a crucial role in the colony-forming abilities of the basal cell population in prostate cancer. IL-4 may therefore contribute to disease relapse by providing a niche that is favourable for the clonogenic growth of prostate cancer stem cells.
Collapse
|
26
|
Frajacomo FTT, de Souza Padilha C, Marinello PC, Guarnier FA, Cecchini R, Duarte JAR, Deminice R. Solid Ehrlich carcinoma reproduces functional and biological characteristics of cancer cachexia. Life Sci 2016; 162:47-53. [DOI: 10.1016/j.lfs.2016.08.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/01/2016] [Accepted: 08/09/2016] [Indexed: 12/23/2022]
|
27
|
Sharmila R, Sindhu G, Arockianathan PM. Nephroprotective effect of β-sitosterol on N-diethylnitrosamine initiated and ferric nitrilotriacetate promoted acute nephrotoxicity in Wistar rats. J Basic Clin Physiol Pharmacol 2016; 27:473-482. [PMID: 26982615 DOI: 10.1515/jbcpp-2015-0085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/09/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND The most abundant plant sterol β-sitosterol is widely used for treating heart diseases and chronic inflammatory conditions. The objective of the current study was to evaluate the nephroprotective effect of β-sitosterol against nephrotoxicants which were studied using renal function markers, antioxidant and lipid peroxidation status, and inflammatory markers. METHODS Male albino Wistar rats were randomly grouped into four: group 1 was vehicle control rats (0.1% carboxymethyl cellulose [CMC]); group 2 was rats treated with N-diethylnitrosamine (DEN) (200 mg/kg body weight [bw] i.p. on the 15th day) and ferric nitrilotriacetate (Fe-NTA) (9 mg/kg bw i.p. on 30th and 32nd days); group 3 was rats that received β-sitosterol (20 mg/kg bw in 0.1% CMC, p.o. for 32 days) 2 weeks prior to the exposure to the nephrotoxicant; and group 4 was rats that received β-sitosterol alone. The experiment was terminated after the 24 h of last dosage of Fe-NTA, and all the animals were sacrificed. The blood, liver and kidney from each group were analyzed for biochemical, molecular and histological changes. RESULTS All the parameters showed significant changes in DEN and Fe-NTA treated animals, whereas β-sitosterol pretreated animals' altered biochemical parameters were restored to near normal. Histopathological and immunoexpression studies on tissues also corroborate the biochemical endpoints. CONCLUSIONS Administration of β-sitosterol to nephrotoxicity induced rats showed significant positive changes in biochemical parameters, histopathological and immunohistochemical observations, and up-regulation of Nrf2 gene expression. From this, it was clear that β-sitosterol showed renal protective function.
Collapse
|
28
|
Lerner L, Tao J, Liu Q, Nicoletti R, Feng B, Krieger B, Mazsa E, Siddiquee Z, Wang R, Huang L, Shen L, Lin J, Vigano A, Chiu MI, Weng Z, Winston W, Weiler S, Gyuris J. MAP3K11/GDF15 axis is a critical driver of cancer cachexia. J Cachexia Sarcopenia Muscle 2016; 7:467-82. [PMID: 27239403 PMCID: PMC4863827 DOI: 10.1002/jcsm.12077] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/16/2015] [Accepted: 09/10/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cancer associated cachexia affects the majority of cancer patients during the course of the disease and thought to be directly responsible for about a quarter of all cancer deaths. Current evidence suggests that a pro-inflammatory state may be associated with this syndrome although the molecular mechanisms responsible for the development of cachexia are poorly understood. The purpose of this work was the identification of key drivers of cancer cachexia that could provide a potential point of intervention for the treatment and/or prevention of this syndrome. METHODS Genetically engineered and xenograft tumour models were used to dissect the molecular mechanisms driving cancer cachexia. Cytokine profiling from the plasma of cachectic and non-cachectic cancer patients and mouse models was utilized to correlate circulating cytokine levels with the cachexia phenotype. RESULTS Utilizing engineered tumour models we identified MAP3K11/GDF15 pathway activation as a potent inducer of cancer cachexia. Increased expression and high circulating levels of GDF15 acted as a key mediator of this process. In animal models, tumour-produced GDF15 was sufficient to trigger the cachexia phenotype. Elevated GDF15 circulating levels correlated with the onset and progression of cachexia in animal models and in patients with cancer. Inhibition of GDF15 biological activity with a specific antibody reversed body weight loss and restored muscle and fat tissue mass in several cachectic animal models regardless of their complex secreted cytokine profile. CONCLUSIONS The combination of correlative observations, gain of function, and loss of function experiments validated GDF15 as a key driver of cancer cachexia and as a potential therapeutic target for the treatment and/or prevention of this syndrome.
Collapse
Affiliation(s)
- Lorena Lerner
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Julie Tao
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Qing Liu
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | | | - Bin Feng
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Brian Krieger
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Elizabeth Mazsa
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Zakir Siddiquee
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Ruoji Wang
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Lucia Huang
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Novartis Institutes for BioMedical Research 211 Massachusetts Ave. Cambridge MA 02139 USA
| | - Luhua Shen
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Moderna Therapeutics 200 Technology Square Cambridge MA 02139 USA
| | - Jie Lin
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Stealth Peptides Inc.275 Grove Street, Ste.3-107 Newton MA 02466 USA
| | - Antonio Vigano
- McGill Nutrition and Performance Laboratory; (MNUPAL) McGill University Health Centre (MUHC) Montreal Canada
| | - M Isabel Chiu
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; Enumeral Biomedical Corp One Kendall Square Building 400 Cambridge MA 02139 USA
| | - Zhigang Weng
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - William Winston
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA; POTENZA Therapeutics 700 Main Street Cambridge MA 02139 USA
| | - Solly Weiler
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| | - Jeno Gyuris
- AVEO Oncology One Broadway 14th Floor Cambridge MA 02142 USA
| |
Collapse
|
29
|
Liu Y, Wang ZX, Cao Y, Zhang G, Chen WB, Jiang CP. Preoperative inflammation-based markers predict early and late recurrence of hepatocellular carcinoma after curative hepatectomy. Hepatobiliary Pancreat Dis Int 2016; 15:266-74. [PMID: 27298102 DOI: 10.1016/s1499-3872(16)60094-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Recurrence of hepatocellular carcinoma (HCC) after curative resection remains a major cause of treatment failure and tumor-related death. Patterns of HCC recurrence can be categorized into early recurrence and late recurrence which have different underlying mechanisms. In this study, we investigated if simple inflammation-based clinical markers can distinguish patterns of recurrence after curative resection of HCC. METHODS A retrospective analysis of 223 patients who underwent curative hepatectomy for HCC was performed. Preoperative inflammation-based factors including neutrophil/lymphocyte ratio (NLR), platelet/lymphocyte ratio, gamma-glutamyl transferase/alanine aminotransferase ratio, aspartate aminotransferase/platelet ratio index (APRI) and prognostic nutritional index together with other clinicopathologic parameters were evaluated by univariate analysis and multivariate analysis to identify independent prognostic factors. By combining risk factors, predictive models were established to distinguish populations at high risk of early or late recurrence. RESULTS Age ≤50 years, resection margin ≤1 cm, TNM stage III-IV, NLR>2.75, APRI>0.23 and positive alpha-fetoprotein were independent adverse prognostic factors for early recurrence. Patients with three or more risk factors were at significant higher risk of early recurrence. APRI>0.23 and positive hepatitis B e antigen (HBeAg) were independent risk factors of late recurrence, the coexistence of high APRI and positive HBeAg increased the risk of late recurrence. CONCLUSIONS Preoperative inflammation-based prognostic factors predict early and late recurrence of HCC after curative resection. Different prognostic factor combinations distinguish high-risk populations of early or late HCC recurrence.
Collapse
Affiliation(s)
- Yue Liu
- Department of Hepatobiliary Surgery, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China.
| | | | | | | | | | | |
Collapse
|
30
|
Belizário JE, Fontes-Oliveira CC, Borges JP, Kashiabara JA, Vannier E. Skeletal muscle wasting and renewal: a pivotal role of myokine IL-6. SPRINGERPLUS 2016; 5:619. [PMID: 27330885 PMCID: PMC4870483 DOI: 10.1186/s40064-016-2197-2] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/20/2016] [Indexed: 12/20/2022]
Abstract
Adult skeletal tissue is composed of heterogeneous population of cells that constantly self-renew by means of a controlled process of activation and proliferation of tissue-resident stem cells named satellite cells. Many growth factors, cytokines and myokines produced by skeletal muscle cells play critical roles in local regulation of the inflammatory process and skeletal muscle regeneration during different pathological conditions. IL-6 is a pleiotropic cytokine released in large amount during infection, autoimmunity and cancer. Low levels of IL-6 can promote activation of satellite cells and myotube regeneration while chronically elevated production promote skeletal muscle wasting. These distinct effects may be explained by a crosstalk of the IL-6/IL-6 receptor and gp130 trans-signaling pathway that oppose to regenerative and anti-inflammatory of the classical IL-6 receptor signaling pathway. Here we discuss on potential therapeutic strategies using monoclonal antibodies to IL-6R for the treatment of skeletal muscle wasting and cachexia. We also highlight on the IL-6/JAK/STAT and FGF/p38αβ MAPK signaling pathways in satellite cell activation and the use of protein kinase inhibitors for tailoring and optimizing satellite cell proliferation during the skeletal muscle renewal. Future investigations on the roles of the IL-6 classical and trans-signaling pathways in both immune and non-immune cells in skeletal muscle tissue will provide new basis for therapeutic approaches to reverse atrophy and degeneration of skeletal muscles in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- José E Belizário
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | | | - Janaina Padua Borges
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | - Janete Akemi Kashiabara
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, Avenida Lineu Prestes, 1524, São Paulo, SP 05508-900 Brazil
| | - Edouard Vannier
- Division of Geographic Medicine and Infectious Disease, Tufts Medical Center, Boston, MA 02111 USA
| |
Collapse
|
31
|
Therapeutic Implications for Overcoming Radiation Resistance in Cancer Therapy. Int J Mol Sci 2015; 16:26880-913. [PMID: 26569225 PMCID: PMC4661850 DOI: 10.3390/ijms161125991] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 09/29/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Ionizing radiation (IR), such as X-rays and gamma (γ)-rays, mediates various forms of cancer cell death such as apoptosis, necrosis, autophagy, mitotic catastrophe, and senescence. Among them, apoptosis and mitotic catastrophe are the main mechanisms of IR action. DNA damage and genomic instability contribute to IR-induced cancer cell death. Although IR therapy may be curative in a number of cancer types, the resistance of cancer cells to radiation remains a major therapeutic problem. In this review, we describe the morphological and molecular aspects of various IR-induced types of cell death. We also discuss cytogenetic variations representative of IR-induced DNA damage and genomic instability. Most importantly, we focus on several pathways and their associated marker proteins responsible for cancer resistance and its therapeutic implications in terms of cancer cell death of various types and characteristics. Finally, we propose radiation-sensitization strategies, such as the modification of fractionation, inflammation, and hypoxia and the combined treatment, that can counteract the resistance of tumors to IR.
Collapse
|
32
|
Jiang F, Zhang Z, Zhang Y, Pan X, Yu L, Liu S. L-Carnitine Ameliorates Cancer Cachexia in Mice Partly via the Carnitine Palmitoyltransferase-Associated PPAR-γ Signaling Pathway. Oncol Res Treat 2015; 38:511-6. [PMID: 26452216 DOI: 10.1159/000439550] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 08/17/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND L-Carnitine has been demonstrated to ameliorate cachectic symptoms. In the present study, we sought to investigate the role of the peroxisome proliferator-activated receptor-γ (PPAR-γ) signaling pathway in the ameliorative effects of L-carnitine on cancer cachexia in a colon-26 tumor-bearing mouse model. METHODS The cachectic mice received L-carnitine (p.o.) or etomoxir (i.p.), or pioglitazone hydrochloride (p.o.) or GW9662 (i.p.). The physiological cachexia parameters, biochemical parameters, and serum cytokines were measured. The expression levels of representative molecules in the PPAR-γ signaling pathway were measured by using quantitative real-time polymerase chain reaction (qRT-PCR) or Western blot analysis. RESULTS Oral administration of L-carnitine at 9 mg/kg/day improved the cachexia parameters and biochemical parameters in cancer cachectic mice. The elevated serum concentrations of interleukin (IL)-6 and tumor necrosis factor-α (TNF-α) were decreased by L-carnitine. These ameliorative effects of L-carnitine were lessened by the carnitine palmitoyltransferase I (CPT I) inhibitor, etomoxir. The mRNA and protein expression levels of PPAR-α and PPAR-γ were decreased in the livers of cancer cachectic mice and increased after L-carnitine administration, which attenuated the increased mRNA expression levels of sterol-regulatory element-binding protein-1c (SREBP-1c) and fatty acid synthase (FAS). Similar to pioglitazone, L-carnitine augmented the phosphorylation of PPAR-γ and attenuated the expression levels of phospho-p65 and cyclooxygenase (COX)-2. Additionally, the above-mentioned effects of L-carnitine were reversed by GW9662. CONCLUSION L-Carnitine exerts its ameliorative effects in cancer cachexia in association with the PPAR-γ signaling pathway.
Collapse
Affiliation(s)
- Fang Jiang
- Department of Gastroenterology, Zhabei District Central Hospital, Shanghai, China
| | | | | | | | | | | |
Collapse
|
33
|
Duan Y, Li Z, Cheng S, Chen Y, Zhang L, He J, Liao Q, Yang L, Gong Z, Sun LQ. Nasopharyngeal carcinoma progression is mediated by EBER-triggered inflammation via the RIG-I pathway. Cancer Lett 2015; 361:67-74. [PMID: 25721089 DOI: 10.1016/j.canlet.2015.02.037] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/14/2015] [Accepted: 02/16/2015] [Indexed: 12/12/2022]
Abstract
EBERs (EBER1 and EBER2) are suggested to be involved in cellular transformation and tumor growth. Cytoplasmic pattern recognition receptor-RIG-I, which is characterized by the recognition of viral dsRNAs, could efficiently trigger the downstream pathways of innate immunity. Although some previous reports have shown that EBERs and RIG-I associate with hematological malignancies, the role of EBERs-RIG-I signaling in solid tumors remains to be clarified. Here we demonstrate that EBER mediation of the inflammatory response via RIG-I contributes to NPC development in vitro and in vivo. We first verified that the expression level of RIG-I was associated with EBER transcription in a dose-dependent manner in NPC cells and specimens from NPC patients. Furthermore, pro-inflammatory cytokine transcription and release were sharply reduced after RIG-I knockdown compared with the control shRNA group in the presence of EBERs, accompanied by an attenuation of the NF-κB and MAPK signaling pathways. Consequently, the tumor burden was greatly alleviated in the RIG-I knockdown group in a xenograft model. In addition, macrophage colony-stimulating factor (M-CSF) and monocyte chemoattractant protein (MCP-1), which promote the maturation and attraction of tumor-associated macrophages, were stimulated upon the introduction of EBERs, and this upregulation conceivably led to the tumor-promoting subset transition of the macrophages. Taken together, our results reveal that EBERs could promote NPC progression through RIG-I-mediated cancer-related inflammation.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Carcinoma
- Cell Differentiation
- Coculture Techniques
- Cytokines/genetics
- Cytokines/metabolism
- DEAD Box Protein 58
- DEAD-box RNA Helicases/antagonists & inhibitors
- DEAD-box RNA Helicases/genetics
- DEAD-box RNA Helicases/metabolism
- Disease Progression
- Female
- Humans
- Immunoenzyme Techniques
- Immunoprecipitation
- Inflammation/immunology
- Inflammation/metabolism
- Inflammation/pathology
- Inflammation Mediators/analysis
- Interferon Regulatory Factor-3/genetics
- Interferon Regulatory Factor-3/metabolism
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Nasopharyngeal Carcinoma
- Nasopharyngeal Neoplasms/immunology
- Nasopharyngeal Neoplasms/metabolism
- Nasopharyngeal Neoplasms/pathology
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- RNA, Viral/antagonists & inhibitors
- RNA, Viral/genetics
- RNA, Viral/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, Immunologic
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yumei Duan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; The department of pathology of Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhi Li
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shiyue Cheng
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Chen
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lu Zhang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiang He
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiong Liao
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lifang Yang
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China; Cancer Research Institute, Central South University, Changsha 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Interleukin-6 (IL-6) has emerged as a cytokine involved in cachexia progression with some cancers. This review will present the recent breakthroughs in animal models and humans related to targeting IL-6 as a cancer cachexia therapy. RECENT FINDINGS IL-6 can target adipose, skeletal muscle, gut, and liver tissue, which can all affect cachectic patient recovery. IL-6 trans-signaling through the soluble IL-6R has the potential to amplify IL-6 signaling in the cachectic patient. In the skeletal muscle, chronic IL-6 exposure induces proteasome and autophagy protein degradation pathways that lead to wasting. IL-6 is also indirectly associated with AMP-activated kinase (AMPK) and nuclear factor kappa B (NF-κB) activation. Several mouse cancer models have clearly demonstrated that blocking IL-6 and associated signaling can attenuate cachexia progression. Additionally, pharmaceuticals targeting IL-6 and associated signaling can relieve some cachectic symptoms in cancer patients. Research with cachectic mice has demonstrated that exercise and nutraceutical administration can interact with chronic IL-6 signaling during cachexia progression. SUMMARY IL-6 remains a promising therapeutic strategy for attenuating cachexia progression with many types of cancer. However, improvement of this treatment will require a better understanding of the indirect and direct effects of IL-6 as well as its tissue-specific actions in the cancer patient.
Collapse
Affiliation(s)
- Aditi A Narsale
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC 29208
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, SC 29208
| | - James A. Carson
- Integrative Muscle Biology Laboratory, Department of Exercise Science, University of South Carolina, Columbia, SC 29208
- Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208
- Division of Applied Physiology, Department of Exercise Science, University of South Carolina, Columbia, SC 29208
| |
Collapse
|
35
|
Ma JD, Heavey SF, Revta C, Roeland EJ. Novel investigational biologics for the treatment of cancer cachexia. Expert Opin Biol Ther 2014; 14:1113-20. [PMID: 24707881 DOI: 10.1517/14712598.2014.907788] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Cancer cachexia is a complex multifactorial syndrome characterized by ongoing, irreversible skeletal muscle loss, leading to progressive functional impairment. Several investigational biologics targeting key inflammatory pathways and/or the myostatin/activin type II receptor pathway are in development. AREAS COVERED Novel therapies include ALD518, MABp1, IP-1510, OHR/AVR118, bimagrumab and REGN1033 and are discussed. For each investigational therapy, the mechanism of action, preclinical data, cachexia definition, indication and clinical data are discussed. EXPERT OPINION A critical look of the study protocols and two key challenges limiting the successful evaluation of these agents include: i) lack of a clinically meaningful cachexia definition; and ii) identification and treatment of cachexia in late stage. We describe our observations and clinical experience in an effort to redirect and promote successful strategies to evaluate these novel investigational biologics.
Collapse
Affiliation(s)
- Joseph D Ma
- University of California (UC), San Diego, Skaggs School of Pharmacy & Pharmaceutical Sciences , 9500 Gilman Drive, MC 0714, La Jolla, CA 92093-0714 , USA
| | | | | | | |
Collapse
|
36
|
Martinez-Outschoorn U, Sotgia F, Lisanti MP. Tumor microenvironment and metabolic synergy in breast cancers: critical importance of mitochondrial fuels and function. Semin Oncol 2014; 41:195-216. [PMID: 24787293 DOI: 10.1053/j.seminoncol.2014.03.002] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metabolic synergy or metabolic coupling between glycolytic stromal cells (Warburg effect) and oxidative cancer cells occurs in human breast cancers and promotes tumor growth. The Warburg effect or aerobic glycolysis is the catabolism of glucose to lactate to obtain adenosine triphosphate (ATP). This review summarizes the main findings on this stromal metabolic phenotype, and the associated signaling pathways, as well as the critical role of oxidative stress and autophagy, all of which promote carcinoma cell mitochondrial metabolism and tumor growth. Loss of Caveolin 1 (Cav-1) and the upregulation of monocarboxylate transporter 4 (MCT4) in stromal cells are novel markers of the Warburg effect and metabolic synergy between stromal and carcinoma cells. MCT4 and Cav-1 are also breast cancer prognostic biomarkers. Reactive oxygen species (ROS) are key mediators of the stromal Warburg effect. High ROS also favors cancer cell mitochondrial metabolism and tumorigenesis, and anti-oxidants can reverse this altered stromal and carcinoma metabolism. A pseudo-hypoxic state with glycolysis and low mitochondrial metabolism in the absence of hypoxia is a common feature in breast cancer. High ROS induces loss of Cav-1 in stromal cells and is sufficient to generate a pseudo-hypoxic state. Loss of Cav-1 in the stroma drives glycolysis and lactate extrusion via HIF-1α stabilization and the upregulation of MCT4. Stromal cells with loss of Cav-1 and/or high expression of MCT4 also show a catabolic phenotype, with enhanced macroautophagy. This catabolic state in stromal cells is driven by hypoxia-inducible factor (HIF)-1α, nuclear factor κB (NFκB), and JNK activation and high ROS generation. A feed-forward loop in stromal cells regulates pseudo-hypoxia and metabolic synergy, with Cav-1, MCT4, HIF-1α, NFκB, and ROS as its key elements. Metabolic synergy also may occur between cancer cells and cells in distant organs from the tumor. Cancer cachexia, which is due to severe organismal metabolic dysregulation in myocytes and adipocytes, shares similarities with stromal-carcinoma metabolic synergy, as well. In summary, metabolic synergy occurs when breast carcinoma cells induce a nutrient-rich microenvironment to promote tumor growth. The process of tumor metabolic synergy is a multistep process, due to the generation of ROS, and the induction of catabolism with autophagy, mitophagy and glycolysis. Studying epithelial-stromal interactions and metabolic synergy is important to better understand the ecology of cancer and the metabolic role of different cell types in tumor progression.
Collapse
Affiliation(s)
| | - Federica Sotgia
- University of Manchester, Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Manchester, United Kingdom
| | - Michael P Lisanti
- University of Manchester, Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Manchester, United Kingdom
| |
Collapse
|
37
|
Martinez-Outschoorn UE, Lisanti MP, Sotgia F. Catabolic cancer-associated fibroblasts transfer energy and biomass to anabolic cancer cells, fueling tumor growth. Semin Cancer Biol 2014; 25:47-60. [PMID: 24486645 DOI: 10.1016/j.semcancer.2014.01.005] [Citation(s) in RCA: 319] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/17/2014] [Accepted: 01/17/2014] [Indexed: 12/22/2022]
Abstract
Fibroblasts are the most abundant "non-cancerous" cells in tumors. However, it remains largely unknown how these cancer-associated fibroblasts (CAFs) promote tumor growth and metastasis, driving chemotherapy resistance and poor clinical outcome. This review summarizes new findings on CAF signaling pathways and their emerging metabolic phenotypes that promote tumor growth. Although it is well established that altered cancer metabolism enhances tumor growth, little is known about the role of fibroblast metabolism in tumor growth. New studies reveal that metabolic coupling occurs between catabolic fibroblasts and anabolic cancer cells, in many types of human tumors, including breast, prostate, and head & neck cancers, as well as lymphomas. These catabolic phenotypes observed in CAFs are secondary to a ROS-induced metabolic stress response. Mechanistically, this occurs via HIF1-alpha and NFκB signaling, driving oxidative stress, autophagy, glycolysis and senescence in stromal fibroblasts. These catabolic CAFs then create a nutrient-rich microenvironment, to metabolically support tumor growth, via the local stromal generation of mitochondrial fuels (lactate, ketone bodies, fatty acids, glutamine, and other amino acids). New biomarkers of this catabolic CAF phenotype (such as caveolin-1 (Cav-1) and MCT4), which are reversible upon treatment with anti-oxidants, are strong predictors of poor clinical outcome in various types of human cancers. How cancer cells metabolically reprogram fibroblasts can also help us to understand the effects of cancer cells at an organismal level, explaining para-neoplastic phenomena, such as cancer cachexia. In conclusion, cancer should be viewed more as a systemic disease, that engages the host-organism in various forms of energy-transfer and metabolic co-operation, across a whole-body "ecosystem".
Collapse
Affiliation(s)
| | - Michael P Lisanti
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, UK.
| | - Federica Sotgia
- Manchester Breast Centre & Breakthrough Breast Cancer Research Unit, Institute of Cancer Sciences, University of Manchester, UK; Manchester Centre for Cellular Metabolism (MCCM), University of Manchester, UK.
| |
Collapse
|