1
|
Zhang L, Bonomi PD. Immune System Disorder and Cancer-Associated Cachexia. Cancers (Basel) 2024; 16:1709. [PMID: 38730660 PMCID: PMC11083538 DOI: 10.3390/cancers16091709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer-associated cachexia (CAC) is a debilitating condition marked by muscle and fat loss, that is unresponsive to nutritional support and contributes significantly to morbidity and mortality in patients with cancer. Immune dysfunction, driven by cytokine imbalance, contributes to CAC progression. This review explores the potential relationship between CAC and anti-cancer immune response in pre-clinical and clinical studies. Pre-clinical studies showcase the involvement of cytokines like IL-1β, IL-6, IL-8, IFN-γ, TNF-α, and TGF-β, in CAC. IL-6 and TNF-α, interacting with muscle and adipose tissues, induce wasting through JAK/STAT and NF-κB pathways. Myeloid-derived suppressor cells (MDSCs) exacerbate CAC by promoting inflammation. Clinical studies confirm elevated pro-inflammatory cytokines (IL-6, IL-8, TNFα) and immune markers like the neutrophil-to-lymphocyte ratio (NLR) in patients with CAC. Thus, immunomodulatory mechanisms involved in CAC may impact the anti-neoplastic immune response. Inhibiting CAC mechanisms could enhance anti-cancer therapies, notably immunotherapy. R-ketorolac, a new immunomodulator, reversed the weight loss and increased survival in mice. Combining these agents with immunotherapy may benefit patients with cancer experiencing CAC. Further research is vital to understand the complex interplay between tumor-induced immune dysregulation and CAC during immunotherapy.
Collapse
Affiliation(s)
| | - Philip D. Bonomi
- Division of Hematology/Oncology, Rush University Medical Center, Chicago, IL 60612, USA;
| |
Collapse
|
2
|
Alim LF, Keane C, Souza-Fonseca-Guimaraes F. Molecular mechanisms of tumour necrosis factor signalling via TNF receptor 1 and TNF receptor 2 in the tumour microenvironment. Curr Opin Immunol 2024; 86:102409. [PMID: 38154421 DOI: 10.1016/j.coi.2023.102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/30/2023]
Abstract
Tumour necrosis factor (TNF) is a primary mediator of inflammatory processes by facilitating cell death, immune cell activation and triggering of inflammation. In the cancer context, research has revealed TNF as a multifaceted cytokine that can be both pro- or anti-tumorigenic depending on what context is observed. We explore the plethora of ways that TNF and its receptors manipulate the functional and phenotypic characteristics in the tumour microenvironment (TME) on both tumour cells and immune cells, promoting either tumour elimination or progression. Here, we discuss the latest cutting-edge TNF-focused biologics currently in clinical translation that modifies the TME to derive greater immune responses and therapeutic outcomes, and further give perspectives on the future of targeting TNF in the context of cancer by emerging technological approaches.
Collapse
Affiliation(s)
- Louisa F Alim
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Colm Keane
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; Princess Alexandra Hospital, Woolloongabba, QLD 4102, Australia
| | | |
Collapse
|
3
|
Chan SH, Kuo WH, Wang LH. SCEL regulates switches between pro-survival and apoptosis of the TNF-α/TNFR1/NF-κB/c-FLIP axis to control lung colonization of triple negative breast cancer. J Biomed Sci 2023; 30:93. [PMID: 38037106 PMCID: PMC10688137 DOI: 10.1186/s12929-023-00986-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/19/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Patients with metastatic triple-negative breast cancer (mTNBC) have a higher probability of developing visceral metastasis within 5 years after the initial diagnosis. Therefore, a deeper understanding of the progression and spread of mTNBC is urgently needed. METHODS The isobaric tag for relative and absolute quantitation (iTRAQ)-based LC-MS/MS proteomic approach was applied to identify novel membrane-associated proteins in the lung-tropic metastatic cells. Public domain datasets were used to assess the clinical relevance of the candidate proteins. Cell-based and mouse models were used for biochemical and functional characterization of the protein molecule Sciellin (SCEL) identified by iTRAQ to elucidate its role and underlying mechanism in promoting lung colonization of TNBC cells. RESULTS The iTRAQ-based LC-MS/MS proteomic approach identified a membrane-associated protein SCEL that was overexpressed in the lung-tropic metastatic cells, and its high expression was significantly correlated with the late-stage TNBC and the shorter survival of the patients. Downregulation of SCEL expression significantly impaired the 3D colony-forming ability but not the migration and invasion ability of the lung colonization (LC) cells. Knockdown of SCEL reduced TNF-α-induced activation of the NF-κB/c-FLIP pro-survival and Akt/Erk1/2 growth signaling pathways in the LC cells. Specifically, knockdown of SCEL expression switched TNF-α-mediated cell survival to the caspase 3-dependent apoptosis. Conversely, ectopic expression of SCEL promoted TNF-α-induced activation of NF-κB/c-FLIP pro-survival and Akt/Erk1/2 pro-growth signaling pathway. The result of co-immunoprecipitation (Co-IP) and GST pull-down assay showed that SCEL could interact with TNFR1 to promote its protein stability. The xenograft mouse model experiments revealed that knockdown of SCEL resulted in increase of caspase-3 activity, and decrease of ki67 and TNFR1 expression as well as increase of tumor-associated macrophages in the metastatic lung lesions. Clinically, SCEL expression was found to be positively correlated with TNFR1 in TNBC tissues. Lastly, we showed that blocking TNF-α-mediated cell survival signaling by adalimumab effectively suppressed the lung colonization of the SCEL-positive, but not the SCEL-downregulated LC cells in the tail-vein injection model. CONCLUSIONS Our findings indicate that SCEL plays an essential role in the metastatic lung colonization of TNBC by promoting the TNF-α/TNFR1/NF-κB/c-FLIP survival and Akt/Erk1/2 proliferation signaling. Thus, SCEL may serve as a biomarker for adalimumab treatment of TNBC patients.
Collapse
Affiliation(s)
- Shih-Hsuan Chan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan.
- Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan.
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, 100, Taiwan
| | - Lu-Hai Wang
- Chinese Medicine Research Center, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan.
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
4
|
Amirkhanzadeh Barandouzi Z, Bruner DW, Miller AH, Paul S, Felger JC, Wommack EC, Higgins KA, Shin DM, Saba NF, Xiao C. Associations of inflammation with neuropsychological symptom cluster in patients with Head and neck cancer: A longitudinal study. Brain Behav Immun Health 2023; 30:100649. [PMID: 37396338 PMCID: PMC10308212 DOI: 10.1016/j.bbih.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/17/2023] [Accepted: 06/03/2023] [Indexed: 07/04/2023] Open
Abstract
Purpose Head and neck cancer (HNC) patients may experience multiple co-occurring neuropsychological symptoms (NPS) cluster, including fatigue, depression, pain, sleep disturbance, and cognitive impairment. While inflammation has been attributed as a key mechanism for some of these symptoms, its association with the NPS as a cluster of symptoms is unknown. Thus, the aim of this study was to examine the association between peripheral inflammation and NPS cluster among HNC patients over cancer treatment (radiotherapy with or without chemotherapy). Methods HNC patients were recruited and followed at pre-treatment, end of treatment, three months and one-year post-treatment. Plasma inflammatory markers, including C-reactive protein (CRP), tumor necrosis factor-alpha (TNFA), soluble tumor necrosis factor receptor-2 (sTNFR2), interleukin-1 beta (IL1-β), interleukin-6 (IL-6), interleukin-10 (IL-10), monocyte chemotactic protein-1 (MCP-1), and interleukin-1 receptor antagonist (IL-1RA) and patient-reported NPS cluster were collected at the 4 time points. Associations between inflammatory markers and the NPS cluster were analyzed using linear mixed-effects models and generalized estimating equations (GEE) models controlling covariates. Results 147 HNC patients were eligible for analysis. 56% of the patients received chemoradiotherapy as treatment. The highest NPS cluster score was reported at the end of treatment, which gradually decreased over time. An increase in inflammatory markers including CRP, sTNFR2, IL-6 and IL-1RA was associated with higher continuous NPS cluster scores (p<0.001, p = 0.003, p<0.001, p<0.001; respectively). GEE further confirmed that patients with at least two moderate symptoms had elevated sTNFR2, IL-6, and IL-1RA (p = 0.017, p = 0.038, p = 0.008; respectively). Notably, this positive association between NPS cluster and inflammatory markers was still significant at one-year post-treatment for CRP (p = 0.001), sTNFR2 (p = 0.006), and IL-1RA (p = 0.043). Conclusions Most HNC patients experienced NPS clusters over time, especially immediately after the end of treatment. Elevated inflammation, as represented by inflammatory markers, was strongly associated with worse NPS cluster over time; this trend was also notable at one-year post-treatment. Our findings suggest that peripheral inflammation plays a pivotal role in the NPS cluster over cancer treatment, including long-term follow-ups. Interventions on reducing peripheral inflammation may contribute to alleviating the NPS cluster in cancer patients.
Collapse
Affiliation(s)
| | - Deborah W. Bruner
- School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Andrew H. Miller
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Sudeshna Paul
- School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Jennifer C. Felger
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Evanthia C. Wommack
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Kristin A. Higgins
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Dong M. Shin
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Nabil F. Saba
- School of Medicine, Emory University, 201 Dowman Dr, Atlanta, GA, 30322, USA
| | - Canhua Xiao
- School of Nursing, Emory University, 1520 Clifton Road NE, Atlanta, GA, 30322, USA
| |
Collapse
|
5
|
Schauer T, Henriksson A, Strandberg E, Lindman H, Berntsen S, Demmelmaier I, Raastad T, Nordin K, Christensen JF. Pre-treatment levels of inflammatory markers and chemotherapy completion rates in patients with early-stage breast cancer. Int J Clin Oncol 2023; 28:89-98. [PMID: 36269530 DOI: 10.1007/s10147-022-02255-0] [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: 05/18/2022] [Accepted: 10/09/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Chemotherapy efficacy is largely dependent on treatment adherence, defined by the relative dose intensity (RDI). Identification of new modifiable risk factors associated with low RDI might improve chemotherapy delivery. Here, we evaluated the association between low RDI and pre-chemotherapy factors, including patient- and treatment-related characteristics and markers of inflammation. METHODS This exploratory analysis assessed data from 267 patients with early-stage breast cancer scheduled to undergo (neo-)adjuvant chemotherapy included in the Physical training and Cancer (Phys-Can) trial. The association between low RDI, defined as < 85%, patient-related (age, body mass index, co-morbid condition, body surface area) and treatment-related factors (cancer stage, receptor status, chemotherapy duration, chemotherapy dose, granulocyte colony-stimulating factor) was investigated. Analyses further included the association between RDI and pre-chemotherapy levels of interleukin (IL)-6, IL-8, IL-10, C-reactive protein (CRP) and Tumor Necrosis Factor-alpha (TNF-α) in 172 patients with available blood samples. RESULTS An RDI of < 85% occurred in 31 patients (12%). Univariable analysis revealed a significant association with a chemotherapy duration above 20 weeks (p < 0.001), chemotherapy dose (p = 0.006), pre-chemotherapy IL-8 (OR 1.61; 95% CI (1.01; 2.58); p = 0.040) and TNF-α (OR 2.2 (1.17; 4.53); p = 0.019). In multivariable analyses, inflammatory cytokines were significant association with low RDI for IL-8 (OR: 1.65 [0.99; 2.69]; p = 0.044) and TNF-α (OR 2.95 [1.41; 7.19]; p = 0.007). CONCLUSIONS This exploratory analysis highlights the association of pre-chemotherapy IL-8 and TNF-α with low RDI of chemotherapy for breast cancer. IL-8 and TNF-α may therefore potentially help to identify patients at risk for experiencing dose reductions. Clinical trial number NCT02473003 (registration: June 16, 2015).
Collapse
Affiliation(s)
- Tim Schauer
- Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Anna Henriksson
- Department of Public Health and Caring Sciences, Uppsala University, Husargatan 3, 751 22, Uppsala, Sweden
| | - Emelie Strandberg
- Department of Public Health and Caring Sciences, Uppsala University, Husargatan 3, 751 22, Uppsala, Sweden
| | - Henrik Lindman
- Department of Oncology, Uppsala University, 751 85, Sjukhusvägen, Uppsala, Sweden
| | - Sveinung Berntsen
- Department of Public Health and Caring Sciences, Uppsala University, Husargatan 3, 751 22, Uppsala, Sweden
- Department of Sport Science and Physical Education, University of Agder, Universitetsveien 25, 4630, Kristiansand, Norway
| | - Ingrid Demmelmaier
- Department of Public Health and Caring Sciences, Uppsala University, Husargatan 3, 751 22, Uppsala, Sweden
- Department of Sport Science and Physical Education, University of Agder, Universitetsveien 25, 4630, Kristiansand, Norway
| | - Truls Raastad
- Department of Sport Science and Physical Education, University of Agder, Universitetsveien 25, 4630, Kristiansand, Norway
- Department of Physical Performance, Norwegian School of Sport Sciences, Sognsveien 220, 0806, Oslo, Norway
| | - Karin Nordin
- Department of Public Health and Caring Sciences, Uppsala University, Husargatan 3, 751 22, Uppsala, Sweden
| | - Jesper F Christensen
- Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
- Institute of Exercise and Biomechanics, University of Southern Denmark, Odense, Denmark
- Digestive Disease Center, Bispebjerg Hospital, Copenhagen, Denmark
| |
Collapse
|
6
|
Impact of radiation therapy on fatigue at 1 year in breast cancer survivors in the prospective multicentre CANcer TOxicity cohort. Eur J Cancer 2022; 177:143-153. [PMID: 36356418 DOI: 10.1016/j.ejca.2022.09.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 01/06/2023]
Abstract
BACKGROUND Fatigue is a common and disabling symptom after breast cancer (BC) treatment, significantly impacting patients' quality of life. We aimed to assess the impact of radiation therapy (RT) modalities on fatigue one year after treatment among patients with early-stage BC. METHODS We used CANTO-RT, a subcohort of CANcer TOxicity (CANTO; NCT01993498), a multicentric nationwide prospective cohort of stages I-III BC treated from 2012 to 2017. Our primary outcome was severe global fatigue 1 year after RT completion (European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-C30 score ≥40/100). The secondary outcomes included severe physical, emotional and cognitive fatigue (European Organization for Research and Treatment of Cancer Quality of Life Questionnaire-FA12). RT-related variables were used as independent variables. Multivariable logistic regression models assessed associations between RT-related variables and fatigue. RESULTS The final analytic cohort included 3295 patients. The prevalence of severe global fatigue 1 year after treatment was 33.3%. Internal mammary chain RT (adjusted odds ratio [OR] 1.48 [95% confidence interval [CI] 1.03-2.13; p = 0.0355]) and normofractionated RT (adjusted OR 1.88 [95% CI 1.06-3.31; p = 0.0298]) were associated with increased odds of severe global fatigue. In addition, there was a significant association between normofractionated RT (adjusted OR 1.849 [95% CI 1.04-3.3; p = 0.0354]) and an increased likelihood of severe physical fatigue. CONCLUSION We found a significant association between internal mammary chain RT (versus No), normofractionated RT (versus hypofractionated RT) and increased likelihood of persistent severe global fatigue. Our data add to the current understanding of treatment-related factors affecting fatigue after BC and could lead to personalised interventions to improve the prevention and management of this disabling symptom.
Collapse
|
7
|
Bennett AN, Huang RX, He Q, Lee NP, Sung WK, Chan KHK. Drug repositioning for esophageal squamous cell carcinoma. Front Genet 2022; 13:991842. [PMID: 36246638 PMCID: PMC9554346 DOI: 10.3389/fgene.2022.991842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Esophageal cancer (EC) remains a significant challenge globally, having the 8th highest incidence and 6th highest mortality worldwide. Esophageal squamous cell carcinoma (ESCC) is the most common form of EC in Asia. Crucially, more than 90% of EC cases in China are ESCC. The high mortality rate of EC is likely due to the limited number of effective therapeutic options. To increase patient survival, novel therapeutic strategies for EC patients must be devised. Unfortunately, the development of novel drugs also presents its own significant challenges as most novel drugs do not make it to market due to lack of efficacy or safety concerns. A more time and cost-effective strategy is to identify existing drugs, that have already been approved for treatment of other diseases, which can be repurposed to treat EC patients, with drug repositioning. This can be achieved by comparing the gene expression profiles of disease-states with the effect on gene-expression by a given drug. In our analysis, we used previously published microarray data and identified 167 differentially expressed genes (DEGs). Using weighted key driver analysis, 39 key driver genes were then identified. These driver genes were then used in Overlap Analysis and Network Analysis in Pharmomics. By extracting drugs common to both analyses, 24 drugs are predicted to demonstrate therapeutic effect in EC patients. Several of which have already been shown to demonstrate a therapeutic effect in EC, most notably Doxorubicin, which is commonly used to treat EC patients, and Ixazomib, which was recently shown to induce apoptosis and supress growth of EC cell lines. Additionally, our analysis predicts multiple psychiatric drugs, including Venlafaxine, as repositioned drugs. This is in line with recent research which suggests that psychiatric drugs should be investigated for use in gastrointestinal cancers such as EC. Our study shows that a drug repositioning approach is a feasible strategy for identifying novel ESCC therapies and can also improve the understanding of the mechanisms underlying the drug targets.
Collapse
Affiliation(s)
- Adam N. Bennett
- Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Rui Xuan Huang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Qian He
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nikki P. Lee
- Department of Surgery, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Wing-Kin Sung
- Department of Computer Sciences, National University of Singapore, Singapore, Singapore
| | - Kei Hang Katie Chan
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Epidemiology, Centre for Global Cardiometabolic Health, Brown University, Providence, RI, United States
| |
Collapse
|
8
|
Ferrara M, Samaden M, Ruggieri E, Vénéreau E. Cancer cachexia as a multiorgan failure: Reconstruction of the crime scene. Front Cell Dev Biol 2022; 10:960341. [PMID: 36158184 PMCID: PMC9493094 DOI: 10.3389/fcell.2022.960341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cachexia is a devastating syndrome associated with the end-stage of several diseases, including cancer, and characterized by body weight loss and severe muscle and adipose tissue wasting. Although different cancer types are affected to diverse extents by cachexia, about 80% of all cancer patients experience this comorbidity, which highly reduces quality of life and response to therapy, and worsens prognosis, accounting for more than 25% of all cancer deaths. Cachexia represents an urgent medical need because, despite several molecular mechanisms have been identified, no effective therapy is currently available for this devastating syndrome. Most studies focus on skeletal muscle, which is indeed the main affected and clinically relevant organ, but cancer cachexia is characterized by a multiorgan failure. In this review, we focus on the current knowledge on the multiple tissues affected by cachexia and on the biomarkers with the attempt to define a chronological pathway, which might be useful for the early identification of patients who will undergo cachexia. Indeed, it is likely that the inefficiency of current therapies might be attributed, at least in part, to their administration in patients at the late stages of cachexia.
Collapse
Affiliation(s)
- Michele Ferrara
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Samaden
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Ruggieri
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emilie Vénéreau
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
9
|
Talbert EE, Guttridge DC. Emerging signaling mediators in the anorexia-cachexia syndrome of cancer. Trends Cancer 2022; 8:397-403. [PMID: 35190301 PMCID: PMC9035074 DOI: 10.1016/j.trecan.2022.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/11/2022]
Abstract
The cachexia syndrome in cancer is characterized by weight loss resulting from the combination of anorexia and atrophy of adipose and skeletal muscle. For decades, inflammatory circulatory factors have been identified to regulate wasting, but inhibitors of these factors have not yielded the same clinical benefit as in animal models. Therefore, additional mediators of cachexia likely regulate this syndrome, and such factors might be more suitable for targeted intervention. We highlight several anorexia-cachexia signaling mediators, including activin A, myostatin, GDF15, and lipocalin-2. We discuss current evidence that these factors associate with cachexia in cancer patients, and summarize translational efforts including essential early-phase clinical trials. We conclude with thoughts on targeted and personalized approaches for future anti-cachexia treatments.
Collapse
Affiliation(s)
- Erin E Talbert
- Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University Iowa, Iowa City, IA 52242, USA
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, and the Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
10
|
Miola A, Dal Porto V, Meda N, Perini G, Solmi M, Sambataro F. Secondary Mania induced by TNF-α inhibitors: A systematic review. Psychiatry Clin Neurosci 2022; 76:15-21. [PMID: 34590391 PMCID: PMC9298409 DOI: 10.1111/pcn.13302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/09/2021] [Accepted: 09/13/2021] [Indexed: 12/01/2022]
Abstract
A growing number of studies support a bidirectional relationship between inflammation and bipolar disorders. Tumor necrosis factor-α (TNF-α) inhibitors have recently attracted interest as potential therapeutic compounds for treating depressive symptoms, but the risk for triggering mood switches in patients with or without bipolar disorders remains controversial. Thus, we conducted a systematic review to study the anti-TNF-α medication-induced manic or hypomanic episodes. PubMed, Scopus, Medline, and Embase databases were screened for a comprehensive literature search from inception until November 2020, using The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Out of the initial 75 references, the screening resulted in the inclusion of four case reports (each describing one patient) and a cohort study (in which 40 patients out of 7600-0.53% - experienced elated mood episodes after infliximab administration). Of these 44 patients, 97.7% experienced a manic episode and 2.3% hypomania. 93.2% of patients had no history of psychiatric disorder or psychotropic treatment. Only 6.8% had a history of psychiatric disorders with the affective spectrum (4.6% dysthymia and 2.3% bipolar disorder). The time of onset of manic or hypomanic symptoms varied across TNF-α inhibitors with an early onset for Infliximab and a later onset for Adalimumab and Etanercept. These findings suggest that medications targeting the TNF-α pathway may trigger a manic episode in patients with or without affective disorders. However, prospective studies are needed to evaluate the relative risk of such side effects and identify the population susceptible to secondary mania.
Collapse
Affiliation(s)
- Alessandro Miola
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy.,Casa di Cura Parco dei Tigli, Padova, Italy
| | | | - Nicola Meda
- Department of Medicine, University of Padova, Padova, Italy
| | - Giulia Perini
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy.,Casa di Cura Parco dei Tigli, Padova, Italy
| | - Marco Solmi
- Department of Psychiatry, University of Ottawa, Ottawa, ON, Canada.,Department of Mental Health, The Ottawa Hospital, Ottawa, ON, Canada.,Clinical Epidemiology Program, Ottawa Hospital Research Institute (OHRI), University of Ottawa, Ottawa, ON, Canada
| | - Fabio Sambataro
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| |
Collapse
|
11
|
Yu C, Zhang T, Shi S, Wei T, Wang Q. Potential biomarkers: differentially expressed proteins of the extrinsic coagulation pathway in plasma samples from patients with depression. Bioengineered 2021; 12:6318-6331. [PMID: 34488523 PMCID: PMC8806736 DOI: 10.1080/21655979.2021.1971037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Depression is a severe disabling psychiatric illness and the pathophysiological mechanisms remain unknown. In previous work, we found the changes in extrinsic coagulation (EC) pathway proteins in depressed patients compared with healthy subjects were significant. In this study, we screened differentially expressed proteins (DEPs) in the EC pathway, and explored the molecular mechanism by constructing a protein-protein interaction (PPI) network. The DEPs of the EC pathwaywere initially screened by isobaric tags for relative and absolute quantification (iTRAQ) in plasma samples obtained from 20 depression patients and 20 healthy controls, and were then identified by Enzyme-linked immunosorbent assays (ELISAs). Ingenuity Pathway Analysis (IPA) software was used to analyse pathway. The differentially expressed genes (DEGs) were identified by analyzing the GSE98793 microarray data from the Gene Expression Omnibus database using the Significance Analysis for Microarrays (SAM, version 4.1) statistical method. Cytoscape version 3.4.0 software was used to construct and visualize PPI networks. The results show that Fibrinogen alpha chain (FGA), Fibrinogen beta chain (FGB), Fibrinogen gamma chain (FGG) and Coagulation factor VII (FVII) were screened in the EC pathway from depression patient samples. FGA, FGB, and FGG were significantly up-regulated, and FVII was down-regulated. Thirteen DEGs related to depression and EC pathways were identified from the microarray database. Among them NF-κB Inhibitor Beta (NFKBIB) and Heat shock protein family B (small) member 1 (HSPB1) were highly correlated with EC pathway. We conclude that EC pathway is associated with depression, which provided clues for the biomarker development and the pathogenesis of depression.
Collapse
Affiliation(s)
- Chunyue Yu
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Teli Zhang
- Department of Pharmacy, The People's Hospital of Daqing, Daqing, China
| | - Shanshan Shi
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Taiming Wei
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| | - Qi Wang
- College of Pharmacy, Harbin Medical University-Daqing, Daqing, China
| |
Collapse
|
12
|
Patzelt L, Junker D, Syväri J, Burian E, Wu M, Prokopchuk O, Nitsche U, Makowski MR, Braren RF, Herzig S, Diaz MB, Karampinos DC. MRI-Determined Psoas Muscle Fat Infiltration Correlates with Severity of Weight Loss during Cancer Cachexia. Cancers (Basel) 2021; 13:cancers13174433. [PMID: 34503243 PMCID: PMC8431175 DOI: 10.3390/cancers13174433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/16/2022] Open
Abstract
PURPOSE To evaluate the suitability of psoas and erector spinae muscle proton density fat fraction (PDFF) and fat volume as biomarkers for monitoring cachexia severity in an oncological cohort, and to evaluate regional variances in muscle parameters over time. METHODS In this prospective study, 58 oncological patients were examined by a 3 T MRI receiving between one and five scans. Muscle volume and PDFF were measured, segmentation masks were divided into proximal, middle and distal muscle section. RESULTS A regional variation of fat distribution in erector spinae muscle at baseline was found (p < 0.01). During follow-ups significant relative change of muscle parameters was observed. Relative maximum change of erector spinae muscle showed a significant regional variation. Correlation testing with age as a covariate revealed significant correlations for baseline psoas fat volume (r = -0.55, p < 0.01) and baseline psoas PDFF (r = -0.52, p = 0.02) with maximum BMI change during the course of the disease. CONCLUSION In erector spinae muscles, a regional variation of fat distribution at baseline and relative maximum change of muscle parameters was observed. Our results indicate that psoas muscle PDFF and fat volume could serve as MRI-determined biomarkers for early risk stratification and disease monitoring regarding progression and severity of weight loss in cancer cachexia.
Collapse
Affiliation(s)
- Lisa Patzelt
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
- Correspondence:
| | - Daniela Junker
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Jan Syväri
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Egon Burian
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Mingming Wu
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Olga Prokopchuk
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (O.P.); (U.N.)
| | - Ulrich Nitsche
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (O.P.); (U.N.)
| | - Marcus R. Makowski
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Rickmer F. Braren
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany; (S.H.); (M.B.D.)
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, 69120 Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University Munich, 81675 Munich, Germany
- Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, 85764 Neuherberg, Germany; (S.H.); (M.B.D.)
- Deutsches Zentrum für Diabetesforschung, 85764 Neuherberg, Germany
| | - Dimitrios C. Karampinos
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (D.J.); (J.S.); (E.B.); (M.W.); (M.R.M.); (R.F.B.); (D.C.K.)
| |
Collapse
|
13
|
Peppas S, Pansieri C, Piovani D, Danese S, Peyrin-Biroulet L, Tsantes AG, Brunetta E, Tsantes AE, Bonovas S. The Brain-Gut Axis: Psychological Functioning and Inflammatory Bowel Diseases. J Clin Med 2021; 10:377. [PMID: 33498197 PMCID: PMC7863941 DOI: 10.3390/jcm10030377] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
The brain-gut axis represents a complex bi-directional system comprising multiple interconnections between the neuroendocrine pathways, the autonomous nervous system and the gastrointestinal tract. Inflammatory bowel disease (IBD), comprising Crohn's disease and ulcerative colitis, is a chronic, relapsing-remitting inflammatory disorder of the gastrointestinal tract with a multifactorial etiology. Depression and anxiety are prevalent among patients with chronic disorders characterized by a strong immune component, such as diabetes mellitus, cancer, multiple sclerosis, rheumatoid arthritis and IBD. Although psychological problems are an important aspect of morbidity and of impaired quality of life in patients with IBD, depression and anxiety continue to be under-diagnosed. There is lack of evidence regarding the exact mechanisms by which depression, anxiety and cognitive dysfunction may occur in these patients, and whether psychological disorders are the result of disease activity or determinants of the IBD occurrence. In this comprehensive review, we summarize the role of the brain-gut axis in the psychological functioning of patients with IBD, and discuss current preclinical and clinical data on the topic and therapeutic strategies potentially useful for the clinical management of these patients. Personalized pathways of psychological supports are needed to improve the quality of life in patients with IBD.
Collapse
Affiliation(s)
- Spyros Peppas
- Department of Gastroenterology, Athens Naval Hospital, 11521 Athens, Greece;
| | - Claudia Pansieri
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (C.P.); (S.D.); (E.B.)
- Humanitas Clinical and Research Center–IRCCS, 20089 Milan, Italy
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (C.P.); (S.D.); (E.B.)
- Humanitas Clinical and Research Center–IRCCS, 20089 Milan, Italy
| | - Silvio Danese
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (C.P.); (S.D.); (E.B.)
- Humanitas Clinical and Research Center–IRCCS, 20089 Milan, Italy
| | - Laurent Peyrin-Biroulet
- Department of Gastroenterology, Inserm U1256 NGERE, Nancy University Hospital, Lorraine University, 54500 Vandoeuvre-les-Nancy, France;
| | - Andreas G. Tsantes
- Attiko Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.G.T.); (A.E.T.)
| | - Enrico Brunetta
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (C.P.); (S.D.); (E.B.)
- Humanitas Clinical and Research Center–IRCCS, 20089 Milan, Italy
| | - Argirios E. Tsantes
- Attiko Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.G.T.); (A.E.T.)
| | - Stefanos Bonovas
- Department of Biomedical Sciences, Humanitas University, 20090 Milan, Italy; (C.P.); (S.D.); (E.B.)
- Humanitas Clinical and Research Center–IRCCS, 20089 Milan, Italy
| |
Collapse
|
14
|
Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol 2021; 18:261-279. [PMID: 33469195 DOI: 10.1038/s41571-020-00459-9] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
The immune system has crucial roles in cancer development and treatment. Whereas adaptive immunity can prevent or constrain cancer through immunosurveillance, innate immunity and inflammation often promote tumorigenesis and malignant progression of nascent cancer. The past decade has witnessed the translation of knowledge derived from preclinical studies of antitumour immunity into clinically effective, approved immunotherapies for cancer. By contrast, the successful implementation of treatments that target cancer-associated inflammation is still awaited. Anti-inflammatory agents have the potential to not only prevent or delay cancer onset but also to improve the efficacy of conventional therapeutics and next-generation immunotherapies. Herein, we review the current clinical advances and experimental findings supporting the utility of an anti-inflammatory approach to the treatment of solid malignancies. Gaining a better mechanistic understanding of the mode of action of anti-inflammatory agents and designing more effective treatment combinations would advance the clinical application of this therapeutic approach.
Collapse
Affiliation(s)
- Jiajie Hou
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
15
|
Kilgour MK, MacPherson S, Zacharias LG, Ellis AE, Sheldon RD, Liu EY, Keyes S, Pauly B, Carleton G, Allard B, Smazynski J, Williams KS, Watson PH, Stagg J, Nelson BH, DeBerardinis RJ, Jones RG, Hamilton PT, Lum JJ. 1-Methylnicotinamide is an immune regulatory metabolite in human ovarian cancer. SCIENCE ADVANCES 2021; 7:eabe1174. [PMID: 33523930 PMCID: PMC7817098 DOI: 10.1126/sciadv.abe1174] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Immune regulatory metabolites are key features of the tumor microenvironment (TME), yet with a few exceptions, their identities remain largely unknown. Here, we profiled tumor and T cells from tumor and ascites of patients with high-grade serous carcinoma (HGSC) to uncover the metabolomes of these distinct TME compartments. Cells within the ascites and tumor had pervasive metabolite differences, with a notable enrichment in 1-methylnicotinamide (MNA) in T cells infiltrating the tumor compared with ascites. Despite the elevated levels of MNA in T cells, the expression of nicotinamide N-methyltransferase, the enzyme that catalyzes the transfer of a methyl group from S-adenosylmethionine to nicotinamide, was restricted to fibroblasts and tumor cells. Functionally, MNA induces T cells to secrete the tumor-promoting cytokine tumor necrosis factor alpha. Thus, TME-derived MNA contributes to the immune modulation of T cells and represents a potential immunotherapy target to treat human cancer.
Collapse
Affiliation(s)
- Marisa K Kilgour
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | - Sarah MacPherson
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | | | - Abigail E Ellis
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Ryan D Sheldon
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Elaine Y Liu
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | - Sarah Keyes
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Brenna Pauly
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Gillian Carleton
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | - Bertrand Allard
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
- Institut du Cancer de Montréal, Québec, Canada
| | - Julian Smazynski
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Peter H Watson
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
- Biobanking and Biospecimen Research Services, Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Québec, Canada
- Faculté de Pharmacie, Université de Montréal, Québec, Canada
- Institut du Cancer de Montréal, Québec, Canada
| | - Brad H Nelson
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Ralph J DeBerardinis
- Children's Research Institute, UT Southwestern, Dallas, TX, USA
- Howard Hughes Medical Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Julian J Lum
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.
- Trev and Joyce Deeley Research Centre, BC Cancer, Victoria, BC, Canada
| |
Collapse
|
16
|
Wyart E, Bindels LB, Mina E, Menga A, Stanga S, Porporato PE. Cachexia, a Systemic Disease beyond Muscle Atrophy. Int J Mol Sci 2020; 21:E8592. [PMID: 33202621 PMCID: PMC7696729 DOI: 10.3390/ijms21228592] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/06/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a complication of dismal prognosis, which often represents the last step of several chronic diseases. For this reason, the comprehension of the molecular drivers of such a condition is crucial for the development of management approaches. Importantly, cachexia is a syndrome affecting various organs, which often results in systemic complications. To date, the majority of the research on cachexia has been focused on skeletal muscle, muscle atrophy being a pivotal cause of weight loss and the major feature associated with the steep reduction in quality of life. Nevertheless, defining the impact of cachexia on other organs is essential to properly comprehend the complexity of such a condition and potentially develop novel therapeutic approaches.
Collapse
Affiliation(s)
- Elisabeth Wyart
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy; (E.W.); (E.M.); (A.M.)
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Erica Mina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy; (E.W.); (E.M.); (A.M.)
| | - Alessio Menga
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy; (E.W.); (E.M.); (A.M.)
| | - Serena Stanga
- Neuroscience Institute Cavalieri Ottolenghi, 10043 Orbassano (TO), Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy;
| | - Paolo E. Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Turin, Italy; (E.W.); (E.M.); (A.M.)
| |
Collapse
|
17
|
Age-related cytokine effects on cancer-related fatigue and quality of life in acute myeloid leukemia. J Geriatr Oncol 2020; 11:402-409. [DOI: 10.1016/j.jgo.2019.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/20/2019] [Accepted: 04/09/2019] [Indexed: 12/22/2022]
|
18
|
Kim KI, Kong M, Lee SH, Lee BJ. The efficacy and safety of Kyung-Ok-Ko on cancer-related fatigue in lung cancer patients: Study protocol for a randomized, patients-assessor blind, placebo-controlled, parallel-group, single-center trial. Medicine (Baltimore) 2019; 98:e17717. [PMID: 31689808 PMCID: PMC6946369 DOI: 10.1097/md.0000000000017717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Cancer-related fatigue (CRF) is a major symptom experienced by lung cancer patients receiving chemotherapy and radiation therapy. Since CRF has a multidimensional influence on cancer patients, they may experience physical weakening, a decline in cognitive function, and depression from emotional consequences. Kyung-Ok-Ko is used for improving fatigue or weak physical constitution. It is known to be effective in immune activation, reducing fatigue, and enhancing cognitive function. Although Kyung-Ok-Ko is clinically used for the treatment of CRF, its efficacy and safety against CRF in lung cancer patients are yet to be studied. Therefore, we aimed to investigate the efficacy and safety of Kyung-Ok-Ko. METHODS This is a randomized, placebo-controlled, patients-assessor blind, parallel-group, single-center clinical trial. Lung cancer patients with CRF, after termination of chemo or radiation therapies, are randomized in a 1:1 ratio to receive either Kyung-Ok-Ko or placebo for 6 weeks. The primary outcome is Brief Fatigue Inventory (BFI). The secondary outcomes include Visual Analog Fatigue Scale (VAFS), Functional Assessment of Cancer Therapy (FACIT) Fatigue scale, Hospital Anxiety Depression Scale (HADS), Montreal Cognitive Assessment Korean version (MoCA-K), and Korean pattern identification questionnaire. Adverse events are evaluated by Common Terminology Criteria for Adverse Events (CTCAE). All outcomes and adverse events are assessed at the baseline, mid-treatment, post-treatment, and at 1-month follow-up. DISCUSSION This study investigates whether Kyung-Ok-Ko can alleviate CRF in lung cancer patients. The results of this study will provide clinical evidence for the application of Kyung-Ok-Ko in the treatment of CRF in lung cancer patients. TRIAL REGISTRATION Korean Clinical Trial Registry (http://cris.nih.go.kr; registration number: KCT000666).Trial status: Currently, participant recruitment is ongoing.
Collapse
Affiliation(s)
- Kwan-Il Kim
- Division of Allergy, Immune and Respiratory System, Department of Internal Medicine, College of Korean Medicine, Kyung Hee University
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University
| | - Moonkyo Kong
- Division of Lung and Head and Neck Oncology, Department of Radiation Oncology, Kyung Hee University Medical Center, Kyung Hee University School of Medicine
| | - Seung Hyeun Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Dongdaemun-gu, Seoul, Republic of Korea
| | - Beom-Joon Lee
- Division of Allergy, Immune and Respiratory System, Department of Internal Medicine, College of Korean Medicine, Kyung Hee University
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University
| |
Collapse
|
19
|
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
|
20
|
Huang LW, Olin RL. Inflammatory biomarkers and patient-reported outcomes in acute myeloid leukemia: Refocusing on older adults. J Geriatr Oncol 2019; 11:395-398. [PMID: 31350219 DOI: 10.1016/j.jgo.2019.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Li-Wen Huang
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 505 Parnassus Ave, Hematology/Oncology office M1286, San Francisco, CA 94143, USA.
| | - Rebecca L Olin
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, 400 Parnassus Ave, Box 0324, San Francisco, CA 94143, USA.
| |
Collapse
|
21
|
Himbert C, Ose J, Lin T, Warby CA, Gigic B, Steindorf K, Schrotz-King P, Abbenhardt-Martin C, Zielske L, Boehm J, Ulrich CM. Inflammation- and angiogenesis-related biomarkers are correlated with cancer-related fatigue in colorectal cancer patients: Results from the ColoCare Study. Eur J Cancer Care (Engl) 2019; 28:e13055. [PMID: 31016796 DOI: 10.1111/ecc.13055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 01/09/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023]
Abstract
Cancer-related fatigue is one of the most common side effects of colorectal cancer treatment and is affected by biomedical factors. We investigated the association of inflammation- and angiogenesis-related biomarkers with cancer-related fatigue. Pre-surgery (baseline) serum samples were obtained from n = 236 newly diagnosed colorectal cancer patients. Meso Scale Discovery assays were performed to measure levels of biomarkers for inflammation and angiogenesis (CRP, SAA, IL-6, IL-8, MCP-1, sICAM-1, sVCAM-1, TNFα, VEGFA and VEGFD). Cancer-related fatigue was assessed with the EORTC QLQ-30 questionnaire at baseline and 6 and 12 months post-surgery. We tested associations using Spearman's partial correlations and logistic regression analyses, adjusting for age, sex and body mass index. sICAM-1 and VEGFD showed a significant positive correlation with cancer-related fatigue at baseline and 6-, and 12-month follow-up (sICAM-1: r = 0.19, p = 0.010; r = 0.24, p = 0.004; r = 0.25, p = 0.006; VEGFD: r = 0.20, p = 0.006; r = 0.15, p = 0.06; r = 0.23, p = 0.01 respectively). Biomarkers of inflammation and angiogenesis measured prior to surgery are associated with cancer-related fatigue in colorectal cancer patients throughout various time points. Our results suggest the involvement of overexpressed sICAM-1 and VEGFD in the development of fatigue.
Collapse
Affiliation(s)
- Caroline Himbert
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Jennifer Ose
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Tengda Lin
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Christy A Warby
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Biljana Gigic
- Department of Surgery, University Clinic of Heidelberg, Heidelberg, Germany
| | - Karen Steindorf
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases, Heidelberg, Germany
| | | | | | - Lin Zielske
- Department of Surgery, University Clinic of Heidelberg, Heidelberg, Germany
| | - Juergen Boehm
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, Population Sciences, Salt Lake City, Utah.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah
| |
Collapse
|
22
|
[Pain and fatigue - a systematic review]. ZEITSCHRIFT FUR PSYCHOSOMATISCHE MEDIZIN UND PSYCHOTHERAPIE 2019; 64:365-379. [PMID: 30829171 DOI: 10.13109/zptm.2018.64.4.365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Pain and fatigue - a systematic review Introduction: Chronic pain and fatigue are symptoms that often occur together and characterize the symptom picture of various diseases. Nevertheless, integrative explanatory approaches have so far received little attention in medical practice. METHODS Based on a systematic literature search in the Embase, Medline, PsychInfo, and CENTRAL databases, we searched for high-quality intervention studies for the simultaneous treatment of pain and fatigue. RESULTS From 1,496 total hits, 158 studies were included in the evaluation. The most commonly studied clinical pictures of the symptomcomplex were tumor catabolism, fibromyalgia, chronic fatigue, rheumatoid arthritis, and osteoarthritis.Current explanations of the symptom complex focus on the activation of proinflammatory microglia in centralized pain syndrome with multiple effects on neurotransmission, neuroendocrinology, neuroplasticity, and the autonomic nervous system. In this model, fatigue can be understood as an evolutionary, meaningful symptom protective of the organism. CONCLUSIONS A deeper understanding of the relationship between centralized pain syndrome and fatigue allows for new explanatory and treatment approaches.
Collapse
|
23
|
Argilés JM, López-Soriano FJ, Stemmler B, Busquets S. Therapeutic strategies against cancer cachexia. Eur J Transl Myol 2019; 29:7960. [PMID: 31019661 PMCID: PMC6460215 DOI: 10.4081/ejtm.2019.7960] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 12/18/2018] [Indexed: 01/06/2023] Open
Abstract
Cancer cachexia has two main components: anorexia and metabolic alterations. The main changes associated with the development of this multi-organic syndrome are glucose intolerance, fat depletion and muscle protein hypercatabolism. The aim of this paper is to review the more recent therapeutic approaches designed to counteract the wasting suffered by the cancer patient with cachexia. Among the most promising approaches we can include the use of ghrelin agonists, beta-blockers, beta-adrenergic agonists, androgen receptor agonists and anti-myostatin peptides. The multi-targeted approach seems essential in these treatments, which should include the combination of both nutritional support, drugs and a suitable program of physical exercise, in order to ameliorate both anorexia and the metabolic changes associated with cachexia. In addition, another very important and crucial aspect to be taken into consideration in the design of clinical trials for the treatment of cancer cachexia is to staging cancer patients in relation with the degree of cachexia, in order to start as early as possible this triple approach in the course of the disease, even before the weight loss can be detected.
Collapse
Affiliation(s)
- Josep M Argilés
- Cancer Research Group, Department of Biochemistry and Molecular Biomedicine, Biology Faculty of the Barcelona University, Barcelona, Spain.,Biomedicine Institute, Barcelona University (IBUB), Barcelona, Spain
| | - Francisco Javier López-Soriano
- Cancer Research Group, Department of Biochemistry and Molecular Biomedicine, Biology Faculty of the Barcelona University, Barcelona, Spain.,Biomedicine Institute, Barcelona University (IBUB), Barcelona, Spain
| | | | - Sílvia Busquets
- Cancer Research Group, Department of Biochemistry and Molecular Biomedicine, Biology Faculty of the Barcelona University, Barcelona, Spain.,Biomedicine Institute, Barcelona University (IBUB), Barcelona, Spain
| |
Collapse
|
24
|
Bower JE. The role of neuro-immune interactions in cancer-related fatigue: Biobehavioral risk factors and mechanisms. Cancer 2019; 125:353-364. [PMID: 30602059 DOI: 10.1002/cncr.31790] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022]
Abstract
Fatigue is a common and distressing symptom in both patients with cancer and cancer survivors. There is substantial variation in the severity and persistence of cancer-related fatigue that may be driven by individual differences in host factors, including characteristics that predate the cancer experience as well as responses to cancer and its treatment. This review examines biobehavioral risk factors linked to fatigue and the mechanisms through which they influence fatigue across the cancer continuum, with a focus on neuro-immune processes. Among psychosocial risk factors, childhood adversity is a strong and consistent predictor of cancer-related fatigue; other risk factors include history of depression, catastrophizing, lack of physical activity, and sleep disturbance, with compelling preliminary evidence for loneliness and trait anxiety. Among biologic systems, initial work suggests that alterations in immune, neuroendocrine, and neural processes are associated with fatigue. The identification of key risk factors and underlying mechanisms is critical for the development and deployment of targeted interventions to reduce the burden of fatigue in the growing population of cancer survivors. Given the multidimensional nature of fatigue, interventions that influence multiple systems may be most effective.
Collapse
Affiliation(s)
- Julienne E Bower
- Department of Psychology, Department of Psychiatry and Biobehavioral Sciences, Cousins Center for Psychoneuroimmunology, and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California
| |
Collapse
|
25
|
Yakovenko A, Cameron M, Trevino JG. Molecular therapeutic strategies targeting pancreatic cancer induced cachexia. World J Gastrointest Surg 2018; 10:95-106. [PMID: 30622678 PMCID: PMC6314860 DOI: 10.4240/wjgs.v10.i9.95] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/01/2018] [Accepted: 11/27/2018] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) induced cachexia is a complex metabolic syndrome associated with significantly increased morbidity and mortality and reduced quality of life. The pathophysiology of cachexia is complex and poorly understood. Many molecular signaling pathways are involved in PC and cachexia. Though our understanding of cancer cachexia is growing, therapeutic options remain limited. Thus, further discovery and investigation of the molecular signaling pathways involved in the pathophysiology of cachexia can be applied to development of targeted therapies. This review focuses on three main pathophysiologic processes implicated in the development and progression of cachexia in PC, as well as their utility in the discovery of novel targeted therapies.
Skeletal muscle wasting is the most prominent pathophysiologic anomaly in cachectic patients and driven by multiple regulatory pathways. Several known molecular pathways that mediate muscle wasting and cachexia include transforming growth factor-beta (TGF-β), myostatin and activin, IGF-1/PI3K/AKT, and JAK-STAT signaling. TGF-β antagonism in cachectic mice reduces skeletal muscle catabolism and weight loss, while improving overall survival. Myostatin/activin inhibition has a great therapeutic potential since it plays an essential role in skeletal muscle regulation. Overexpression of insulin-like growth factor binding protein-3 (IGFBP-3) leads to increased ubiquitination associated proteolysis, inhibition of myogenesis, and decreased muscle mass in PC induced cachexia. IGFBP-3 antagonism alleviates muscle cell wasting.
Another component of cachexia is profound systemic inflammation driven by pro-cachectic cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), and interferon gamma (INF-γ). IL-6 antagonism has been shown to reduce inflammation, reduce skeletal muscle loss, and ameliorate cachexia. While TNF-α inhibitors are clinically available, blocking TNF-α signaling is not effective in the treatment of cancer cachexia. Blocking the synthesis or action of acute phase reactants and cytokines is a feasible therapeutic strategy, but no anti-cytokine therapies are currently approved for use in PC. Metabolic alterations such as increased energy expenditure and gluconeogenesis, insulin resistance, fat tissue browning, excessive oxidative stress, and proteolysis with amino acid mobilization support tumor growth and the development of cachexia. Current innovative nutritional strategies for cachexia management include ketogenic diet, utilization of natural compounds such as silibinin, and supplementation with ω3-polyunsaturated fatty acids. Elevated ketone bodies exhibit an anticancer and anticachectic effect. Silibinin has been shown to inhibit growth of PC cells, induce metabolic alterations, and reduce myofiber degradation. Consumption of ω3-polyunsaturated fatty acids has been shown to significantly decrease resting energy expenditure and regulate metabolic dysfunction.
Collapse
Affiliation(s)
- Anastasiya Yakovenko
- University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | - Miles Cameron
- University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | - Jose Gilberto Trevino
- Department of Surgery, University of Florida Health Sciences Center, Gainesville, Florida 32610, United States
| |
Collapse
|
26
|
Herlofson K, Heijnen CJ, Lange J, Alves G, Tysnes OB, Friedman JH, Fagundes CP. Inflammation and fatigue in early, untreated Parkinson's Disease. Acta Neurol Scand 2018; 138:394-399. [PMID: 29947088 DOI: 10.1111/ane.12977] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Parkinson's disease (PD)-related fatigue is a significant clinical problem, and the pathological processes that cause fatigue remain unknown. The aim of the present study was to explore the possible association of peripheral inflammation markers and fatigue in PD. MATERIALS & METHODS We included 47 drug naïve, newly diagnosed PD patients with low (≤3.0) or high (>5.5) fatigue levels as evaluated by the Fatigue Severity Scale (FSS). Strict diagnostic criteria were applied for inclusion. Patients with possible confounding causes for fatigue were excluded. Serum concentrations of a panel of inflammatory markers (IL-8, TNF-α, MCP1, MIP-1β, IL-6, IL-6R, p-selectin, E-selectin-1, ICAM, VCAM-1, CCL5, IL1-Ra, and TNFR1) were measured using ELISA technology in PD patients with and without fatigue to assess the potential relationships of fatigue in newly diagnosed, treatment-naïve patients. RESULTS Fatigued PD patients had significantly higher levels of the IL-1 receptor antagonist (IL1-Ra) (1790 pg/mL (SD1007) vs 1262 pg/mL (SD379)) and of the adhesion molecule VCAM 1 (1071 ng/mL (SD276) vs 895 ng/mL (SD229)) than non-fatigued patients. A binary logistic regression model, including high or low FSS score as the dependent variable and UPDRS motor score, MADRS, MMSE, ESS, and IL1-Ra/VCAM-1 as independent variables, showed a significant effect both for IL1-Ra and VCAM-1. CONCLUSIONS Higher serum levels of the inflammatory molecules IL1-Ra and VCAM-1 were associated with higher fatigue levels in patients with newly diagnosed, drug-naïve PD. These findings highlight an altered immune response as a potential contributor to PD-related fatigue, from the earliest clinical stages of the disease.
Collapse
Affiliation(s)
- K. Herlofson
- Department of Neurology; Sorlandet Hospital; Arendal Norway
| | - C. J. Heijnen
- Department of Symptom Research; M. D. Anderson Cancer Center; Houston TX USA
| | - J. Lange
- The Norwegian Centre for Movement Disorders; Stavanger University Hospital; Stavanger Norway
| | - G. Alves
- The Norwegian Centre for Movement Disorders; Stavanger University Hospital; Stavanger Norway
- Department of Mathematics and Natural Sciences; University of Stavanger; Stavanger Norway
| | - O.-B. Tysnes
- Department of Neurology; Haukeland University Hospital; Department of Clinical Medicine; Bergen Norway
- Department of Clinical Medicine; Haukeland University Hospital; Bergen Norway
| | - J. H. Friedman
- Butler Hospital; Warren Alpert Medical School of Brown University; Providence RI USA
| | - C. P. Fagundes
- Department of Psychology; Rice University and MD Anderson Cancer Center; Houston TX USA
- Department of Behavioral Science; Rice University and MD Anderson Cancer Center; Houston TX USA
| |
Collapse
|
27
|
Emerging role of extracellular vesicles in mediating cancer cachexia. Biochem Soc Trans 2018; 46:1129-1136. [PMID: 30242118 DOI: 10.1042/bst20180213] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/23/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022]
Abstract
Cancer cachexia is a multifactorial metabolic syndrome characterized by the rapid loss of skeletal muscle mass with or without the loss of fat mass. Nearly 50-80% of all cancer patients' experience rapid weight loss results in ∼20% of cancer-related deaths. The levels of pro-inflammatory and pro-cachectic factors were significantly up-regulated in cachexia patients when compared with the patients who were without cachexia. It is becoming evident that these factors work synergistically to induce cancer cachexia. Extracellular vesicles (EVs) including exosomes and microvesicles are implicated in cell-cell communication, immune response, tissue repair, epigenetic regulation, and in various diseases including cancer. It has been reported that these EVs regulate cancer progression, metastasis, organotropism and chemoresistance. In recent times, the role of EVs in regulating cancer cachexia is beginning to unravel. The aim of this mini article is to review the recent knowledge gained in the field of EVs and cancer cachexia. Specifically, the role of tumour cell-derived EVs in promoting catabolism in distally located skeletal muscles and adipose tissue will be discussed.
Collapse
|
28
|
Cui W, Ning Y, Hong W, Wang J, Liu Z, Li MD. Crosstalk Between Inflammation and Glutamate System in Depression: Signaling Pathway and Molecular Biomarkers for Ketamine's Antidepressant Effect. Mol Neurobiol 2018; 56:3484-3500. [PMID: 30140973 DOI: 10.1007/s12035-018-1306-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/07/2018] [Indexed: 12/25/2022]
Abstract
Depression is a worldwide illness with a significant impact on both family and society. Conventional antidepressants are ineffective for more than 30% of patients. In such patients, who have what is called treatment-resistant depression (TRD), inflammatory biomarkers are expressed excessively in both the central nervous system (CNS) and the peripheral blood. Ketamine, a glutamate receptor antagonist, exerts a rapid and sustained therapeutic effect in patients with TRD. Thus, the investigation of the relations between inflammation and glutamate underlying depression has drawn great attention. Inflammation influences glutamate release, transmission, and metabolism, resulting in accumulated extracellular glutamate in the CNS. Downstream of the glutamate receptors, the mammalian target of rapamycin (mTOR) signaling pathway plays a key role in mediating ketamine's antidepressant effect by improving neurogenesis and plasticity. Based on the mechanism and clinical evidence of the inflammatory contribution to the pathogenesis of depression, extensive research has been devoted to inflammatory biomarkers of the clinical response of depression to ketamine. The inconsistent findings from the biomarker investigations are at least partially attributable to the heterogeneity of depression, limited sample size, and complex gene-environment interactions. Deep exploration of the clinical observations and the underlying mechanism of ketamine's antidepressant response can provide new insights into the selection of specific groups of depressed patients for ketamine treatment and to aid in monitoring the therapeutic effect during antidepressant medication. Further, targeting persistent inflammation in patients with TRD and the key molecules mediating ketamine's antidepressant effect may encourage the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Wenyan Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yuping Ning
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wu Hong
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ju Wang
- School of Biomedical Engineering, Tianjin Medical University, Tianjin, China
| | - Zhening Liu
- The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ming D Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, China. .,Research Center for Air Pollution and Health, Zhejiang University, Hangzhou, China. .,Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ, USA.
| |
Collapse
|
29
|
Rolvering C, Zimmer AD, Ginolhac A, Margue C, Kirchmeyer M, Servais F, Hermanns HM, Hergovits S, Nazarov PV, Nicot N, Kreis S, Haan S, Behrmann I, Haan C. The PD-L1- and IL6-mediated dampening of the IL27/STAT1 anticancer responses are prevented by α-PD-L1 or α-IL6 antibodies. J Leukoc Biol 2018; 104:969-985. [PMID: 30040142 DOI: 10.1002/jlb.ma1217-495r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/15/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin-27 (IL27) is a type-I cytokine of the IL6/IL12 family and is predominantly secreted by activated macrophages and dendritic cells. We show that IL27 induces STAT factor phosphorylation in cancerous cell lines of different tissue origin. IL27 leads to STAT1 phosphorylation and recapitulates an IFN-γ-like response in the microarray analyses, with up-regulation of genes involved in antiviral defense, antigen presentation, and immune suppression. Like IFN-γ, IL27 leads to an up-regulation of TAP2 and MHC-I proteins, which mediate increased tumor immune clearance. However, both cytokines also upregulate proteins such as PD-L1 (CD274) and IDO-1, which are associated with immune escape of cancer. Interestingly, differential expression of these genes was observed within the different cell lines and when comparing IL27 to IFN-γ. In coculture experiments of hepatocellular carcinoma (HCC) cells with peripheral blood mononuclear cells, pre-treatment of the HCC cells with IL27 resulted in lowered IL2 production by anti-CD3/-CD28 activated T-lymphocytes. Addition of anti-PD-L1 antibody, however, restored IL2 secretion. The levels of other TH 1 cytokines were also enhanced or restored upon administration of anti-PD-L1. In addition, we show that the suppression of IL27 signaling by IL6-type cytokine pre-stimulation-mimicking a situation occurring, for example, in IL6-secreting tumors or in tumor inflammation-induced cachexia-can be antagonized by antibodies against IL6-type cytokines or their receptors. Therapeutically, the antitumor effects of IL27 (mediated, e.g., by increased antigen presentation) might thus be increased by combining IL27 with blocking antibodies against PD-L1 or/and IL6-type cytokines.
Collapse
Affiliation(s)
- Catherine Rolvering
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Andreas D Zimmer
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Aurélien Ginolhac
- University of Luxembourg, Life Sciences Research Unit-Bioinformatics Core Facility, Belvaux, Luxembourg
| | - Christiane Margue
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Mélanie Kirchmeyer
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Florence Servais
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Heike M Hermanns
- University Hospital Würzburg, Medical Clinic II, Division of Hepatology, Würzburg, Germany
| | - Sabine Hergovits
- University Hospital Würzburg, Medical Clinic II, Division of Hepatology, Würzburg, Germany
| | - Petr V Nazarov
- Proteome and Genome Research Unit, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Nathalie Nicot
- Proteome and Genome Research Unit, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Stephanie Kreis
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Serge Haan
- University of Luxembourg, Life Sciences Research Unit-Molecular Disease Mechanisms Laboratory, Belvaux, Luxembourg
| | - Iris Behrmann
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| | - Claude Haan
- University of Luxembourg, Life Sciences Research Unit-Signal Transduction Laboratory, Belvaux, Luxembourg
| |
Collapse
|
30
|
Jordan KR, Loman BR, Bailey MT, Pyter LM. Gut microbiota-immune-brain interactions in chemotherapy-associated behavioral comorbidities. Cancer 2018; 124:3990-3999. [PMID: 29975400 DOI: 10.1002/cncr.31584] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 04/06/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Increasing scientific attention is focused on the gut-brain axis, including the ability of the gastrointestinal (GI) tract to modulate central nervous system function. Changes in the intestinal microbiome can influence affective-like behavior, cognitive performance, fatigue, and sleep in rodents and humans. Patients with cancer who are receiving chemotherapy experience similar negative behavioral changes and concurrent GI symptoms. These chemotherapy comorbidities can be long-lasting and may reduce patients' quality of life and motivation to comply with treatment. This review summarizes the clinical and preclinical evidence supporting a role for the intestinal microbiome in mediating behavioral comorbidities through peripheral immune activation in patients with cancer who are receiving chemotherapy. In addition, evidence suggesting that targeted modification of the intestinal microbiome during cancer treatment could ameliorate associated behavioral comorbidities is reviewed.
Collapse
Affiliation(s)
- Kelley R Jordan
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio.,Department of Neuroscience, The Ohio State University, Columbus, Ohio
| | - Brett R Loman
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Michael T Bailey
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio.,Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leah M Pyter
- The Institute for Behavioral Medicine Research, The Ohio State University, Columbus, Ohio.,Department of Neuroscience, The Ohio State University, Columbus, Ohio.,Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, Ohio
| |
Collapse
|
31
|
Santos JC, Pyter LM. Neuroimmunology of Behavioral Comorbidities Associated With Cancer and Cancer Treatments. Front Immunol 2018; 9:1195. [PMID: 29930550 PMCID: PMC6001368 DOI: 10.3389/fimmu.2018.01195] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/14/2018] [Indexed: 12/27/2022] Open
Abstract
Behavioral comorbidities (depression, anxiety, fatigue, cognitive disturbances, and neuropathic pain) are prevalent in cancer patients and survivors. These mental and neurological health issues reduce quality-of-life, which is a significant societal concern given the increasing rates of long-term survival after various cancers. Hypothesized causes of behavioral comorbidities with cancer include tumor biology, stress associated with the cancer experience, and cancer treatments. A relatively recent leading mechanism by which these causes contribute to changes in neurobiology that underlie behavior is inflammation. Indeed, both basic and clinical research indicates that peripheral inflammation leads to central inflammation and behavioral changes in other illness contexts. Given the limitations of assessing neuroimmunology in clinical populations, this review primarily synthesizes evidence of neuroimmune and neuroinflammatory changes due to two components of cancer (tumor biology and cancer treatments) that are associated with altered affective-like or cognitive behaviors in rodents. Specifically, alterations in microglia, neuroinflammation, and immune trafficking to the brain are compiled in models of tumors, chemotherapy, and/or radiation. Evidence-based neuronal mechanisms by which these neuroimmune changes may lead to changes in behavior are proposed. Finally, converging evidence in clinical cancer populations is discussed.
Collapse
Affiliation(s)
- Jessica C Santos
- Department of Basic and Applied Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Leah M Pyter
- Departments of Psychiatry and Behavioral Health and Neuroscience, The Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
32
|
Tomlinson D, Robinson P, Oberoi S, Cataudella D, Culos-Reed N, Davis H, Duong N, Gibson F, Götte M, Hinds P, Nijhof S, van der Torre P, Cabral S, Dupuis L, Sung L. Pharmacologic interventions for fatigue in cancer and transplantation: a meta-analysis. Curr Oncol 2018; 25:e152-e167. [PMID: 29719440 PMCID: PMC5927795 DOI: 10.3747/co.25.3883] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Our objective was to determine whether, compared with control interventions, pharmacologic interventions reduce the severity of fatigue in patients with cancer or recipients of hematopoietic stem-cell transplantation (hsct). Methods For a systematic review, we searched medline, embase, the Cochrane Central Register of Controlled Trials, cinahl, and Psychinfo for randomized trials of systemic pharmacologic interventions for the management of fatigue in patients with cancer or recipients of hsct. Two authors independently identified studies and abstracted data. Methodologic quality was assessed using the Cochrane Risk of Bias tool. The primary outcome was fatigue severity measured using various fatigue scales. Data were synthesized using random-effects models. Results In the 117 included trials (19,819 patients), the pharmacologic agents used were erythropoietins (n = 31), stimulants (n = 19), l-carnitine (n = 6), corticosteroids (n = 5), antidepressants (n = 5), appetite stimulants (n = 3), and other agents (n = 48). Fatigue was significantly reduced with erythropoietin [standardized mean difference (smd): -0.52; 95% confidence interval (ci): -0.89 to -0.14] and with methylphenidate (smd: -0.36; 95% ci: -0.56 to -0.15); modafinil (or armodafinil) and corticosteroids were not effective. Conclusions Erythropoietin and methylphenidate significantly reduced fatigue severity in patients with cancer and in recipients of hsct. Concerns about the safety of those agents might limit their usefulness. Future research should identify effective interventions for fatigue that have minimal adverse effects.
Collapse
Affiliation(s)
- D. Tomlinson
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON
| | | | - S. Oberoi
- Pediatric Oncology Group of Ontario, Toronto, ON
| | - D. Cataudella
- Department of Pediatric Psychology, Children’s Hospital, London Health Sciences Centre, London, ON
| | - N. Culos-Reed
- Faculty of Kinesiology, University of Calgary, Calgary, AB
| | - H. Davis
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON
| | - N. Duong
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON
| | - F. Gibson
- Centre for Outcomes and Experiences Research in Children’s Health, Illness, and Disability, Great Ormond Street Hospital for Children NHS Foundation Trust, London, and School of Health Sciences, University of Surrey, Guildford, U.K
| | - M. Götte
- University Hospital Essen, Center for Child and Adolescent Medicine, Department of Pediatric Hematology/Oncology, Essen, Germany
| | - P. Hinds
- Department of Nursing Science, Professional Practice, and Quality, Children’s National Health System; and Department of Pediatrics, George Washington University, Washington, DC, U.S.A
| | - S.L. Nijhof
- Division of Pediatrics, Wilhelmina Children’s Hospital (part of UMC Utrecht), Utrecht, Netherlands
| | - P. van der Torre
- Division of Pediatrics, Wilhelmina Children’s Hospital (part of UMC Utrecht), Utrecht, Netherlands
| | - S. Cabral
- Pediatric Oncology Group of Ontario, Toronto, ON
| | - L.L. Dupuis
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON
- Department of Pharmacy, The Hospital for Sick Children; and Leslie Dan Faculty of Pharmacy, University of Toronto, The Hospital for Sick Children, Toronto, ON
| | - L. Sung
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON
| |
Collapse
|
33
|
Ji YB, Bo CL, Xue XJ, Weng EM, Gao GC, Dai BB, Ding KW, Xu CP. Association of Inflammatory Cytokines With the Symptom Cluster of Pain, Fatigue, Depression, and Sleep Disturbance in Chinese Patients With Cancer. J Pain Symptom Manage 2017; 54:843-852. [PMID: 28797869 DOI: 10.1016/j.jpainsymman.2017.05.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/30/2017] [Accepted: 05/25/2017] [Indexed: 02/01/2023]
Abstract
CONTEXT Pain, fatigue, depression, and sleep disturbance are common in patients with cancer and usually co-occur as a symptom cluster. However, the mechanism underlying this symptom cluster is unclear. OBJECTIVES This study aimed to identify subgroups of cluster symptoms, compare demographic and clinical characteristics between subgroups, and examine the associations between inflammatory cytokines and cluster symptoms. METHODS Participants were 170 Chinese inpatients with cancer from two tertiary hospitals. Inflammatory markers including interleukin-6 (IL-6), interleukin-1 receptor antagonist, and tumor necrosis factor alpha were measured. Intergroup differences and associations of inflammatory cytokines with the cluster symptoms were examined with one-way analyses of variance and logistic regression. RESULTS Based on cluster analysis, participants were categorized into Subgroup 1 (all low symptoms), Subgroup 2 (low pain and moderate fatigue), or Subgroup 3 (moderate-to-high on all symptoms). The three subgroups differed significantly in Eastern Cooperative Oncology Group (ECOG) performance status, sex, residence, current treatment, education, economic status, and inflammatory cytokines levels (all P < 0.05). Compared with Subgroup 1, Subgroup 3 had a significantly poorer ECOG physical performance status and higher IL-6 levels, were more often treated with combined chemoradiotherapy, and were more likely to be rural residents. IL-6 and ECOG physical performance status were significantly associated with 1.246-fold (95% CI 1.114-1.396) and 31.831-fold (95% CI 6.017-168.385) increased risk of Subgroup 3. CONCLUSION Our findings suggest that IL-6 levels are associated with cluster symptoms in cancer patients. Clinicians should identify patients at risk for more severe symptoms and formulate novel target interventions to improve symptom management.
Collapse
Affiliation(s)
- Yan-Bo Ji
- Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China
| | - Chun-Lu Bo
- School of Nursing, Cheeloo Health Science Center, Shandong University, Jinan, Shandong Province, China
| | - Xiu-Juan Xue
- Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China
| | - En-Ming Weng
- Yanggu No. 2 People's Hospital, Liaocheng, Shandong Province, China
| | - Guang-Chao Gao
- School of Nursing, Taishan Medical College, Tai'an, Shandong Province, China
| | - Bei-Bei Dai
- School of Nursing, Taishan Medical College, Tai'an, Shandong Province, China
| | - Kai-Wen Ding
- School of Nursing, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Cui-Ping Xu
- Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
34
|
Pérez-De-Lis M, Retamozo S, Flores-Chávez A, Kostov B, Perez-Alvarez R, Brito-Zerón P, Ramos-Casals M. Autoimmune diseases induced by biological agents. A review of 12,731 cases (BIOGEAS Registry). Expert Opin Drug Saf 2017; 16:1255-1271. [DOI: 10.1080/14740338.2017.1372421] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Marta Pérez-De-Lis
- Servicio de Anestesiologia y Reanimación, Complejo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Soledad Retamozo
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Hospital Privado Universitario de Córdoba, Córdoba, Argentina
- Instituto De Investigaciones En Ciencias De La Salud (INICSA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Alejandra Flores-Chávez
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Unidad de Investigación Biomédica 02, Unidad de Investigación en Epidemiología Clínica, Centro Médico Nacional de Occidente (CMNO), Instituto Mexicano del Seguro Social (IMSS), Hospital de Especialidades, Guadalajara, Mexico
- Programa de Doctorado en Ciencias Médicas, Centro Universitario de Investigaciones Biomédicas (CUIB), Universidad de Colima, Colima, Mexico
| | | | | | - Pilar Brito-Zerón
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Autoimmune Diseases Unit, Department of Medicine, Hospital CIMA- Sanitas, Barcelona, Spain
| | - Manuel Ramos-Casals
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, Hospital Clínic, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| |
Collapse
|
35
|
Novel targeted therapies for cancer cachexia. Biochem J 2017; 474:2663-2678. [PMID: 28751550 DOI: 10.1042/bcj20170032] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 02/06/2023]
Abstract
Anorexia and metabolic alterations are the main components of the cachectic syndrome. Glucose intolerance, fat depletion, muscle protein catabolism and other alterations are involved in the development of cancer cachexia, a multi-organ syndrome. Nutritional approach strategies are not satisfactory in reversing the cachectic syndrome. The aim of the present review is to deal with the recent therapeutic targeted approaches that have been designed to fight and counteract wasting in cancer patients. Indeed, some promising targeted therapeutic approaches include ghrelin agonists, selective androgen receptor agonists, β-blockers and antimyostatin peptides. However, a multi-targeted approach seems absolutely essential to treat patients affected by cancer cachexia. This approach should not only involve combinations of drugs but also nutrition and an adequate program of physical exercise, factors that may lead to a synergy, essential to overcome the syndrome. This may efficiently reverse the metabolic changes described above and, at the same time, ameliorate the anorexia. Defining this therapeutic combination of drugs/nutrients/exercise is an exciting project that will stimulate many scientific efforts. Other aspects that will, no doubt, be very important for successful treatment of cancer wasting will be an optimized design of future clinical trials, together with a protocol for staging cancer patients in relation to their degree of cachexia. This will permit that nutritional/metabolic/pharmacological support can be started early in the course of the disease, before severe weight loss occurs. Indeed, timing is crucial and has to be taken very seriously when applying the therapeutic approach.
Collapse
|
36
|
McSorley ST, Dolan RD, Roxburgh CSD, McMillan DC, Horgan PG. How and why systemic inflammation worsens quality of life in patients with advanced cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.1080/23809000.2017.1331705] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Stephen T. McSorley
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow, UK
| | - Ross D. Dolan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow, UK
| | | | - Donald C. McMillan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow, UK
| | - Paul G. Horgan
- Academic Unit of Surgery, School of Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
37
|
|
38
|
Georgin-Lavialle S, Moura DS, Salvador A, Chauvet-Gelinier JC, Launay JM, Damaj G, Côté F, Soucié E, Chandesris MO, Barète S, Grandpeix-Guyodo C, Bachmeyer C, Alyanakian MA, Aouba A, Lortholary O, Dubreuil P, Teyssier JR, Trojak B, Haffen E, Vandel P, Bonin B, Hermine O, Gaillard R. Mast cells' involvement in inflammation pathways linked to depression: evidence in mastocytosis. Mol Psychiatry 2016; 21:1511-1516. [PMID: 26809839 DOI: 10.1038/mp.2015.216] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 10/24/2015] [Accepted: 11/24/2015] [Indexed: 12/17/2022]
Abstract
Converging sources of evidence point to a role for inflammation in the development of depression, fatigue and cognitive dysfunction. More precisely, the tryptophan (TRP) catabolism is thought to play a major role in inflammation-induced depression. Mastocytosis is a rare disease in which chronic symptoms, including depression, are related to mast cell accumulation and activation. Our objectives were to study the correlations between neuropsychiatric features and the TRP catabolism pathway in mastocytosis in order to demonstrate mast cells' potential involvement in inflammation-induced depression. Fifty-four patients with mastocytosis and a mean age of 50.1 years were enrolled in the study and compared healthy age-matched controls. Depression and stress were evaluated with the Beck Depression Inventory revised and the Perceived Stress Scale. All patients had measurements of TRP, serotonin (5-HT), kynurenine (KYN), indoleamine 2,3-dioxygenase 1 (IDO1) activity (ratio KYN/TRP), kynurenic acid (KA) and quinolinic acid (QA). Patients displayed significantly lower levels of TRP and 5-HT without hypoalbuminemia or malabsorption, higher IDO1 activity, and higher levels of KA and QA, with an imbalance towards the latter. High perceived stress and high depression scores were associated with low TRP and high IDO1 activity. In conclusion, TRP metabolism is altered in mastocytosis and correlates with perceived stress and depression, demonstrating mast cells' involvement in inflammation pathways linked to depression.
Collapse
Affiliation(s)
- S Georgin-Lavialle
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - D S Moura
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Laboratoire de Psychopathologie et Processus de Santé, EA 4057, IUPDP, Institut de Psychologie, Université Paris Descartes, Paris, France
| | - A Salvador
- Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France
| | - J-C Chauvet-Gelinier
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | - J-M Launay
- Laboratoire de biochimie et biologie moléculaire, Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - G Damaj
- Service des Maladies du Sang, Centre Hospitalier Universitaire, Hôpital Sud, Amiens, France
| | - F Côté
- INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - E Soucié
- INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - M-O Chandesris
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - S Barète
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France
| | - C Grandpeix-Guyodo
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France
| | - C Bachmeyer
- Service de médecine Interne, Hôpital Tenon, Université Pierre et Marie Curie, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - M-A Alyanakian
- Laboratoire d'immunologie, Hôpital Necker, Paris, France
| | - A Aouba
- Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - O Lortholary
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Service des infectieuses et tropicales, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants malades, Paris, France
| | - P Dubreuil
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM UMR 891, Centre de Recherche en Cancérologie de Marseille, Laboratoire d'Hématopoïèse Moléculaire et Fonctionnelle, Marseille, France
| | - J-R Teyssier
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France
| | - B Trojak
- Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Service de Psychiatrie, Département de Neurosciences, Dijon, France
| | - E Haffen
- Laboratoire de génétique, CHU, PTB, 2 rue Angélique Ducoudray, Dijon, France.,Service de Psychiatrie, CHU de Besançon, Besançon, France.,Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France
| | - P Vandel
- Laboratoire de Neurosciences EA 481, IFR 133, Université of Franche-Comté, Besançon, France.,Centre d'Investigation Clinique CIC-IT 808 INSERM, CHU de Besaçon, Besançon, France
| | - B Bonin
- Service de Psychiatrie, Département de Neurosciences, Dijon, France.,Laboratoire de Psychologie et Psychopathologie Médicale (IFR 100), Université de Bourgogne, Dijon, France
| | | | - O Hermine
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,INSERM U1163 and CNRS ERL 8254 and Laboratory of Physiopathology and Treatment of Hematological Disorders, Hôpital Necker-Enfants malades, Paris, France.,Service d'Hématologie Adulte, Université Paris Descartes, Sorbonne, Paris Cité, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Hôpital Necker-Enfants malades, Paris, France
| | - R Gaillard
- Centre de référence des mastocytoses, Université Paris Descartes, Sorbonne, Paris Cité, Hôpital Necker Enfants malades, Fondation Imagine, Paris, France.,Laboratoire de "Physiopathologie des maladies Psychiatriques", Centre de Psychiatrie et Neurosciences U894, INSERM, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Psychiatrie, Centre Hospitalier Sainte-Anne, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine Paris Descartes, Paris, France.,Human Histopathology and Animal Models, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| |
Collapse
|
39
|
Brown PJ, Rutherford BR, Yaffe K, Tandler JM, Ray JL, Pott E, Chung S, Roose SP. The Depressed Frail Phenotype: The Clinical Manifestation of Increased Biological Aging. Am J Geriatr Psychiatry 2016; 24:1084-1094. [PMID: 27618646 PMCID: PMC5069140 DOI: 10.1016/j.jagp.2016.06.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 06/14/2016] [Accepted: 06/16/2016] [Indexed: 01/06/2023]
Abstract
Depression in later life is a severe public health problem, associated with higher rates of mortality, suicide, and dementia. Effectiveness of treatment is limited by the failure to deconstruct the heterogeneity of the illness and because diagnostic criteria, pathophysiological models, and treatment algorithms for depression are primarily based on studies of younger adults even though symptoms of the illness and physiology of the patient change with age. Thus, understanding how aging interacts with depressive illness may elucidate endophenotypes of late-life depression with different clinical manifestations and underlying mechanisms that can then be targeted with more personalized approaches to treatment. This paper proposes a model for the critical confluence between depression and frailty, a high-risk morbidity and mortality syndrome of later life. This model hypothesizes that characteristics of frailty in adults with late life depression represent the clinical manifestation of greater biological aging and their presence in the context of a depressive illness exposes elders to deleterious trajectories. Potential common biological substrates that may result in the manifestation of the depressed frail phenotype including mitochondrial functioning, dopaminergic neurotransmission, and inflammatory processes and implications for the assessment and treatment of adults with late-life depression are discussed. As society continues to live longer, the preservation of the quality of these added years becomes paramount, and the combined impact of depression and frailty on the preservation of this quality warrants the attention of clinical researchers and physicians.
Collapse
Affiliation(s)
- Patrick J. Brown
- College of Physicians and Surgeons, Columbia University, New York, NY USA,New York State Psychiatric Institute, New York, NY USA
| | - Bret R. Rutherford
- College of Physicians and Surgeons, Columbia University, New York, NY USA,New York State Psychiatric Institute, New York, NY USA
| | - Kristine Yaffe
- Neurology, Psychiatry, Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | | | | | - Emily Pott
- New York State Psychiatric Institute, New York, NY USA
| | - Sarah Chung
- New York State Psychiatric Institute, New York, NY USA
| | - Steven P. Roose
- College of Physicians and Surgeons, Columbia University, New York, NY USA,New York State Psychiatric Institute, New York, NY USA
| |
Collapse
|
40
|
Haroon E, Fleischer CC, Felger JC, Chen X, Woolwine BJ, Patel T, Hu XP, Miller AH. Conceptual convergence: increased inflammation is associated with increased basal ganglia glutamate in patients with major depression. Mol Psychiatry 2016; 21:1351-7. [PMID: 26754953 PMCID: PMC4940313 DOI: 10.1038/mp.2015.206] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/08/2015] [Accepted: 10/30/2015] [Indexed: 01/19/2023]
Abstract
Inflammation and altered glutamate metabolism are two pathways implicated in the pathophysiology of depression. Interestingly, these pathways may be linked given that administration of inflammatory cytokines such as interferon-α to otherwise non-depressed controls increased glutamate in the basal ganglia and dorsal anterior cingulate cortex (dACC) as measured by magnetic resonance spectroscopy (MRS). Whether increased inflammation is associated with increased glutamate among patients with major depression is unknown. Accordingly, we conducted a cross-sectional study of 50 medication-free, depressed outpatients using single-voxel MRS, to measure absolute glutamate concentrations in basal ganglia and dACC. Multivoxel chemical shift imaging (CSI) was used to explore creatine-normalized measures of other metabolites in basal ganglia. Plasma and cerebrospinal fluid (CSF) inflammatory markers were assessed along with anhedonia and psychomotor speed. Increased log plasma C-reactive protein (CRP) was significantly associated with increased log left basal ganglia glutamate controlling for age, sex, race, body mass index, smoking status and depression severity. In turn, log left basal ganglia glutamate was associated with anhedonia and psychomotor slowing measured by the finger-tapping test, simple reaction time task and the Digit Symbol Substitution Task. Plasma CRP was not associated with dACC glutamate. Plasma and CSF CRP were also associated with CSI measures of basal ganglia glutamate and the glial marker myoinositol. These data indicate that increased inflammation in major depression may lead to increased glutamate in the basal ganglia in association with glial dysfunction and suggest that therapeutic strategies targeting glutamate may be preferentially effective in depressed patients with increased inflammation as measured by CRP.
Collapse
Affiliation(s)
- E Haroon
- Emory Behavioral Immunology Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Emory Biomedical Imaging Technology Center, Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Winship Cancer Center, Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - C C Fleischer
- Emory Biomedical Imaging Technology Center, Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - J C Felger
- Emory Behavioral Immunology Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Center, Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - X Chen
- Emory Biomedical Imaging Technology Center, Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - B J Woolwine
- Emory Behavioral Immunology Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Center, Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - T Patel
- Department of Anesthesiology, Emory University School of Medicine, Atlanta, GA, USA
| | - X P Hu
- Emory Biomedical Imaging Technology Center, Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - A H Miller
- Emory Behavioral Immunology Program, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Center, Department of Hematology and Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
41
|
Fielitz J. Cancer cachexia-when proteasomal inhibition is not enough. J Cachexia Sarcopenia Muscle 2016; 7:239-45. [PMID: 27386167 PMCID: PMC4929817 DOI: 10.1002/jcsm.12124] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/29/2016] [Indexed: 01/06/2023] Open
Affiliation(s)
- Jens Fielitz
- Department of Molecular Cardiology, Experimental and Clinical Research Center (ECRC) Charité--Universitätsmedizin Berlin, Max Delbrück Center (MDC) for Molecular Medicine in the Helmholtz Association Berlin Germany; Department of Cardiology Heart Center Brandenburg and Medical School Brandenburg (MHB) Bernau Germany
| |
Collapse
|
42
|
Aapro M, Scotte F, Bouillet T, Currow D, Vigano A. A Practical Approach to Fatigue Management in Colorectal Cancer. Clin Colorectal Cancer 2016; 16:275-285. [PMID: 29066018 DOI: 10.1016/j.clcc.2016.04.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 04/08/2016] [Accepted: 04/27/2016] [Indexed: 01/06/2023]
Abstract
Cancer-related fatigue is serious and complex, as well as one of the most common symptoms experienced by patients with colorectal cancer, with the potential to compromise quality of life, activities of daily living, and ultimately survival. There is a lack of consensus about the definition of cancer-related fatigue; however, definitions have been put forward by the European Association for Palliative Care (EAPC) and the National Comprehensive Cancer Network (NCCN). Numerous cancer- and treatment-related factors can contribute to fatigue, including disease progression, comorbidities, medical complications such as anemia, side effects of other medications, and a number of physical and psychologic factors. This underlines the importance of tackling factors that may contribute to fatigue before reducing the dose of treatment. NCCN guidelines and the EAPC have proposed approaches to managing fatigue in cancer patients; however, relatively few therapeutic agents have been demonstrated to reduce fatigue in randomized controlled trials. It is recognized that physical activity produces many beneficial physiologic modifications to markers of physical performance that can help to counteract various causes of fatigue. In appropriately managed and monitored patients with colorectal cancer, emerging evidence indicates that exercise programs may have a favorable influence on cancer-related fatigue, quality of life, and clinical outcomes, and therefore may help patients tolerate chemotherapy. This review assesses fatigue in patients with colorectal cancer and proposes updates to a treatment algorithm that may help clinicians manage this common problem.
Collapse
Affiliation(s)
- Matti Aapro
- Multidisciplinary Oncology Institute, Clinique de Genolier, Genolier, Switzerland.
| | - Florian Scotte
- Oncology Department, Georges Pompidou European Hospital, Paris, France
| | - Thierry Bouillet
- Oncology Department, University Hospital Avicenne, Bobigny, France
| | - David Currow
- Palliative and Supportive Services, Flinders University, Adelaide, Australia
| | - Antonio Vigano
- McGill Nutrition and Performance Laboratory and Division of Supportive and Palliative Care, McGill University Health Centre, Montreal, Canada
| |
Collapse
|
43
|
Asher A, Fu JB, Bailey C, Hughes JK. Fatigue among patients with brain tumors. CNS Oncol 2016; 5:91-100. [PMID: 26987038 PMCID: PMC6047436 DOI: 10.2217/cns-2015-0008] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/14/2016] [Indexed: 12/27/2022] Open
Abstract
Fatigue is a ubiquitous and an extremely distressing symptom among patients with brain tumors (BT), particularly those with high-grade gliomas. The pathophysiology of cancer-related fatigue (CRF) in the context of patients with BT is multifactorial and complex, involving biological, behavioral, medical and social factors. The etiology of CRF in the general oncology population is pointing to the role of inflammatory cytokines as a key factor in the genesis of CRF, but this research is currently limited in the setting of BT. CRF should be screened, assessed and managed according to clinical practice guidelines. Fatigue has recently emerged as a strong, independent prognostic factor for survival that provides incremental prognostic value to the traditional markers of prognosis in recurrent high-grade gliomas. Therefore, strategies to treat fatigue warrant investigation, not only to improve the QOL of a group of patients with often limited life expectancy, but also possibly to optimize survival.
Collapse
Affiliation(s)
- Arash Asher
- Cancer Survivorship & Rehabilitation, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Assistant Clinical Professor, Health Sciences, UCLA, 8700 Beverly Boulevard, AC 1109 Los Angeles, 90048, USA
| | - Jack B Fu
- Department of Palliative, Rehabilitation & Integrative Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1414, Houston, TX 77030, USA
| | - Charlotte Bailey
- Samuel Oschin Comprehensive Cancer Institute at Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA
| | - Jennifer K Hughes
- Department of Rehabilitation, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
44
|
Porporato PE. Understanding cachexia as a cancer metabolism syndrome. Oncogenesis 2016; 5:e200. [PMID: 26900952 PMCID: PMC5154342 DOI: 10.1038/oncsis.2016.3] [Citation(s) in RCA: 356] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 12/05/2015] [Accepted: 12/13/2015] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming occurs in tumors to foster cancer cell proliferation, survival and metastasis, but as well at a systemic level affecting the whole organism, eventually leading to cancer cachexia. Indeed, as cancer cells rely on external sources of nitrogen and carbon skeleton to grow, systemic metabolic deregulation promoting tissue wasting and metabolites mobilization ultimately supports tumor growth. Cachectic patients experience a wide range of symptoms affecting several organ functions such as muscle, liver, brain, immune system and heart, collectively decreasing patients' quality of life and worsening their prognosis. Moreover, cachexia is estimated to be the direct cause of at least 20% of cancer deaths. The main aspect of cachexia syndrome is the unstoppable skeletal muscle and fat storage wasting, even with an adequate caloric intake, resulting in nutrient mobilization – both directly as lipid and amino acids and indirectly as glucose derived from the exploitation of liver gluconeogenesis – that reaches the tumor through the bloodstream. From a metabolic standpoint, cachectic host develops a wide range of dysfunctions, from increased insulin and IGF-1 resistance to induction of mitochondrial uncoupling proteins and fat tissue browning resulting in an increased energy expenditure and heat generation, even at rest. For a long time, cachexia has been merely considered an epiphenomenon of end-stage tumors. However, in specific tumor types, such as pancreatic cancers, it is now clear that patients present markers of tissue wasting at a stage in which tumor is not yet clinically detectable, and that host amino acid supply is required for tumor growth. Indeed, tumor cells actively promote tissue wasting by secreting specific factors such as parathyroid hormone-related protein and micro RNAs. Understanding the molecular and metabolic mediators of cachexia will not only advance therapeutic approaches against cancer, but also improve patients' quality of life.
Collapse
Affiliation(s)
- P E Porporato
- Pole of Pharmacology, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| |
Collapse
|
45
|
Felger JC. The Role of Dopamine in Inflammation-Associated Depression: Mechanisms and Therapeutic Implications. Curr Top Behav Neurosci 2016; 31:199-219. [PMID: 27225499 DOI: 10.1007/7854_2016_13] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Studies investigating the impact of a variety of inflammatory stimuli on the brain and behavior have consistently reported evidence that inflammatory cytokines affect the basal ganglia and dopamine to mediate depressive symptoms related to motivation and motor activity. Findings have included inflammation-associated reductions in ventral striatal responses to hedonic reward, decreased dopamine and dopamine metabolites in cerebrospinal fluid, and decreased availability of striatal dopamine, all of which correlate with symptoms of anhedonia, fatigue, and psychomotor retardation. Similar relationships between alterations in dopamine-relevant corticostriatal reward circuitry and symptoms of anhedonia and psychomotor slowing have also been observed in patients with major depression who exhibit increased peripheral cytokines and other inflammatory markers, such as C-reactive protein. Of note, these inflammation-associated depressive symptoms are often difficult to treat in patients with medical illnesses or major depression. Furthermore, a wealth of literature suggests that inflammation can decrease dopamine synthesis, packaging, and release, thus sabotaging or circumventing the efficacy of standard antidepressant treatments. Herein, the mechanisms by which inflammation and cytokines affect dopamine neurotransmission are discussed, which may provide novel insights into treatment of inflammation-related behavioral symptoms that contribute to an inflammatory malaise.
Collapse
Affiliation(s)
- Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences and The Winship Cancer Institute, Emory University School of Medicine, 1365-B Clifton Road, 5th Floor, Atlanta, GA, 30322, USA.
| |
Collapse
|
46
|
Felger JC, Haroon E, Woolwine BJ, Raison CL, Miller AH. Interferon-alpha-induced inflammation is associated with reduced glucocorticoid negative feedback sensitivity and depression in patients with hepatitis C virus. Physiol Behav 2015; 166:14-21. [PMID: 26703235 DOI: 10.1016/j.physbeh.2015.12.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/23/2015] [Accepted: 12/14/2015] [Indexed: 12/22/2022]
Abstract
Major medical illnesses are associated with increased risk for depression and alterations in hypothalamic-pituitary-adrenal (HPA) axis function. Pathophysiological processes such as inflammation that occur as a part of medical illnesses and their treatments have been shown to cause depressive symptoms, and may also affect the HPA axis. We previously reported that patients with hepatitis C virus chronically administered interferon (IFN)-alpha develop increased evening plasma cortisol concentrations and a flattened diurnal cortisol slope, which correlated with increased tumor necrosis factor (TNF) and its soluble receptor 2 (sTNFR2). Increased TNF and sTNFR2 were further correlated with depression and fatigue scores. The current study examined whether flattened cortisol slope might be secondary to reduced glucocorticoid receptor (GR) sensitivity, by measuring glucocorticoid negative feedback to dexamethasone (DEX) administration followed by corticotropin releasing hormone (CRH) challenge. In an exploratory analysis, 28 male and female patients with hepatitis C virus were studied at baseline (Visit 1) and after 12weeks (Visit 2) of either IFN-alpha plus ribavirin (n=17) or no treatment (n=11). Patients underwent dexamethasone DEX-CRH challenge, neuropsychiatric assessments, and measurement of plasma TNF and sTNFR2 during each visit. IFN-alpha did not affect neuroendocrine responses following CRH but did increase post-DEX cortisol, which was correlated with flattening of the diurnal cortisol slope (r=0.57, p=0.002) and with increased depression scores (r=0.38, p=0.047). Furthermore, the change in post-DEX cortisol was associated with IFN-alpha-induced increase in sTNFR2 (r=0.51, p=006), which was in turn correlated with depression (r=0.63, p<0.001) and fatigue (r=0.51, p=0.005) scores. Whereas the relationship between sTNFR2 and depression scores were independent of the change in post-DEX cortisol, the correlation between post-DEX cortisol and depression scores was not significant when controlling for sTNFR2. These findings suggest that inflammation induced in patients with hepatitis C virus during IFN-alpha therapy precipitates decreased GR sensitivity to lead to a flattened diurnal cortisol slope. Decreased GR sensitivity may in turn further increase inflammation and its ultimate effects on behavior. Treatments that target inflammation and/or GR sensitivity may reduce depressive symptoms associated with medical illnesses.
Collapse
Affiliation(s)
- Jennifer C Felger
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States; The Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States.
| | - Ebrahim Haroon
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States; The Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| | - Bobbi J Woolwine
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States
| | - Charles L Raison
- School of Human Ecology, University of Wisconsin-Madison, Madison, WI 53706, United States; Department of Psychiatry, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Andrew H Miller
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, United States; The Winship Cancer Institute, Emory University, Atlanta, GA 30322, United States
| |
Collapse
|
47
|
Abstract
Depression and inflammation fuel one another. Inflammation plays a key role in depression's pathogenesis for a subset of depressed individuals; depression also primes larger cytokine responses to stressors and pathogens that do not appear to habituate. Accordingly, treatment decisions may be informed by attention to questions of how (pathways) and for whom (predispositions) these links exist, which are the focus of this article. When combined with predisposing factors (moderators such as childhood adversity and obesity), stressors and pathogens can lead to exaggerated or prolonged inflammatory responses. The resulting sickness behaviors (e.g., pain, disturbed sleep), depressive symptoms, and negative health behaviors (e.g., poor diet, a sedentary lifestyle) may act as mediating pathways that lead to further, unrestrained inflammation and depression. Depression, childhood adversity, stressors, and diet can all influence the gut microbiome and promote intestinal permeability, another pathway to enhanced inflammatory responses. Larger, more frequent, or more prolonged inflammatory responses could have negative mental and physical health consequences. In clinical practice, inflammation provides a guide to potential targets for symptom management by signaling responsiveness to certain therapeutic strategies. For example, a theme across research with cytokine antagonists, omega-3 fatty acids, celecoxib, and exercise is that anti-inflammatory interventions have a substantially greater impact on mood in individuals with heightened inflammation. Thus, when inflammation and depression co-occur, treating them in tandem may enhance recovery and reduce the risk of recurrence. The bidirectional links between depression, inflammation, and disease suggest that effective depression treatments could have a far-reaching impact on mood, inflammation, and health.
Collapse
|
48
|
Sergeeva M, Rech J, Schett G, Hess A. Response to peripheral immune stimulation within the brain: magnetic resonance imaging perspective of treatment success. Arthritis Res Ther 2015; 17:268. [PMID: 26477946 PMCID: PMC4610054 DOI: 10.1186/s13075-015-0783-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chronic peripheral inflammation in diseases such as rheumatoid arthritis leads to alterations in central pain processing and consequently to mood disorders resulting from sensitization within the central nervous system and enhanced vulnerability of the medial pain pathway. Proinflammatory cytokines such as tumor necrosis factor (TNF) alpha play an important role herein, and therapies targeting their signaling (i.e., anti-TNF therapies) have been proven to achieve good results. However, the phenomenon of rapid improvement in the patients’ subjective feeling after the start of TNFα neutralization remained confusing, because it was observed long before any detectable signs of inflammation decline. Functional magnetic resonance imaging (fMRI), enabling visualization of brain activity upon peripheral immune stimulation with anti-TNF, has helped to clarify this discrepancy. Moreover, fMRI appeared to work as a reliable tool for predicting prospective success of anti-TNF therapy, which is valuable considering the side effects of the drugs and the high therapy costs. This review, which is mainly guided by neuroimaging studies of the brain, summarizes the state-of-the-art knowledge about communication between the immune system and the brain and its impact on subjective well-being, addresses in more detail the outcome of the abovementioned anti-TNF fMRI studies (rapid response to TNFα blockade within the brain pain matrix and differences in brain activation patterns between prospective therapy responders and nonresponders), and discusses possible mechanisms for the latter phenomena and the predictive power of fMRI.
Collapse
Affiliation(s)
- Marina Sergeeva
- Institut for Experimental Pharmacology, Friedrich Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054, Erlangen, Germany.
| | - Jürgen Rech
- Department of Internal Medicine III, Friedrich Alexander University Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany.
| | - Georg Schett
- Department of Internal Medicine III, Friedrich Alexander University Erlangen-Nürnberg, Ulmenweg 18, 91054, Erlangen, Germany.
| | - Andreas Hess
- Institut for Experimental Pharmacology, Friedrich Alexander University Erlangen-Nürnberg, Fahrstrasse 17, 91054, Erlangen, Germany.
| |
Collapse
|
49
|
Talbert EE, Guttridge DC. Impaired regeneration: A role for the muscle microenvironment in cancer cachexia. Semin Cell Dev Biol 2015; 54:82-91. [PMID: 26385617 DOI: 10.1016/j.semcdb.2015.09.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 09/11/2015] [Indexed: 12/17/2022]
Abstract
While changes in muscle protein synthesis and degradation have long been known to contribute to muscle wasting, a body of literature has arisen which suggests that regulation of the satellite cell and its ensuing regenerative program are impaired in atrophied muscle. Lessons learned from cancer cachexia suggest that this regulation is simply not a consequence, but a contributing factor to the wasting process. In addition to satellite cells, evidence from mouse models of cancer cachexia also suggests that non-satellite progenitor cells from the muscle microenvironment are also involved. This chapter in the series reviews the evidence of dysfunctional muscle repair in multiple wasting conditions. Potential mechanisms for this dysfunctional regeneration are discussed, particularly in the context of cancer cachexia.
Collapse
Affiliation(s)
- Erin E Talbert
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, and the Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Denis C Guttridge
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, and the Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
50
|
Mitchell SA, Hoffman AJ, Clark JC, DeGennaro RM, Poirier P, Robinson CB, Weisbrod BL. Putting evidence into practice: an update of evidence-based interventions for cancer-related fatigue during and following treatment. Clin J Oncol Nurs 2015; 18 Suppl:38-58. [PMID: 25427608 DOI: 10.1188/14.cjon.s3.38-58] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cancer-related fatigue (CRF) has deleterious effects on physical, social, cognitive, and vocational functioning, and causes emotional and spiritual distress for patients and their families; however, it remains under-recognized and undertreated. This article critically reviews and integrates the available empirical evidence supporting the efficacy of pharmacologic and nonpharmacologic treatment approaches to CRF, highlighting new evidence since 2007 and 2009 Putting Evidence Into Practice publications. Interventions that are recommended for practice or likely to be effective in improving fatigue outcomes include exercise; screening for treatable risk factors; management of concurrent symptoms; yoga; structured rehabilitation; Wisconsin ginseng; cognitive-behavioral therapies for insomnia, pain, and depression; mindfulness-based stress reduction; and psychoeducational interventions such as anticipatory guidance, psychosocial support, and energy conservation and activity management. This information can be applied to improve the management of CRF, inform health policy and program development, shape the design of clinical trials of new therapies for CRF, and drive basic and translational research.
Collapse
Affiliation(s)
- Sandra A Mitchell
- Division of Cancer Control and Population Sciences, National Cancer Center, Bethesda, MD
| | - Amy J Hoffman
- College of Nursing, Michigan State University, East Lansing
| | - Jane C Clark
- Georgia Center for Oncology Research and Education in Atlanta
| | | | | | | | | |
Collapse
|