1
|
Yuan L, Li XY, Xu L, Quan SJ, Huang YB, Zheng H. Effects of olanzapine in the improvement of body weight and appetite in patients with cancer or receiving chemotherapy: a systematic review and meta-analysis. Eur J Clin Pharmacol 2025; 81:45-63. [PMID: 39467862 DOI: 10.1007/s00228-024-03770-x] [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: 05/30/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024]
Abstract
OBJECTIVE We aimed to assess the effects of olanzapine in the improvement of body weight and appetite in patients with cancer or receiving chemotherapy through a systematic review and meta-analysis. METHODS We searched the following databases from their inception to April 23, 2024: Embase, PubMed, Web of Science, and the Cochrane Library. The mean difference (MD) and risk ratios were used to calculate by random effects models. The primary outcome was the proportion of patients with > 5% weight gain. RESULTS Seventeen studies with 3457 participants were included. For primary outcomes, 1 study with 124 participants showed olanzapine increased the proportion of patients with > 5% weight gain compared with placebo (60% vs. 9%, P < 0.001). Versus active controls (3 studies, 439 participants), no significant difference in the proportion of patients with > 5% weight gain (RR = 1.69, 95%CI: 0.91 to 3.13, I2 = 27%, P = 0.10), with moderate-quality evidence. Olanzapine increased appetite scores compared to both placebo (1 study, 112 participants; MD = 3, 95%CI: 2.3 to 3.7, P < 0.001) and active controls (2 studies, 106 participants; MD = 4.96, 95%CI: 4.61 to 5.30, I2 = 0%, P < 0.01). For mean weight change, olanzapine showed no significant differences versus placebo (2 studies, 164 participants, MD = 2.78 kg, 95%CI: - 1.60 to 7.17, I2 = 48%, P = 0.21) or active controls (2 studies, 480 participants, MD = 0.44 kg, 95%CI: - 1.04 to 1.91, I2 = 58%, P = 0.56). CONCLUSIONS Olanzapine appears to be a potential option for improving appetite and weight gain in cancer patients. Future trials need to focus on the optimal target dose and use durations of olanzapine. Registered: https://archive.org/details/osf-registrations-kpv4h-v .
Collapse
Affiliation(s)
- Lu Yuan
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin-Yu Li
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Xu
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Si-Jie Quan
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan-Bing Huang
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Zheng
- The Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
2
|
Pranzini E, Muccillo L, Nesi I, Santi A, Mancini C, Lori G, Genovese M, Lottini T, Comito G, Caselli A, Arcangeli A, Sabatino L, Colantuoni V, Taddei ML, Cirri P, Paoli P. Limiting serine availability during tumor progression promotes muscle wasting in cancer cachexia. Cell Death Discov 2024; 10:510. [PMID: 39706853 DOI: 10.1038/s41420-024-02271-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/06/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of body weight occurring in about 80% of cancer patients, frequently representing the leading cause of death. Dietary intervention is emerging as a promising therapeutic strategy to counteract cancer-induced wasting. Serine is the second most-consumed amino acid (AA) by cancer cells and has emerged to be strictly necessary to preserve skeletal muscle structure and functionality. Here, we demonstrate that decreased serine availability during tumor progression promotes myotubes diameter reduction in vitro and induces muscle wasting in in vivo mice models. By investigating the metabolic crosstalk between colorectal cancer cells and muscle cells, we found that incubating myotubes with conditioned media from tumor cells relying on exogenous serine consumption triggers pronounced myotubes diameter reduction. Accordingly, culturing myotubes in a serine-free medium induces fibers width reduction and suppresses the activation of the AKT-mTORC1 pathway with consequent impairment in protein synthesis, increased protein degradation, and enhanced expression of the muscle atrophy-related genes Atrogin1 and MuRF1. In addition, serine-starved conditions affect myoblast differentiation and mitochondrial oxidative metabolism, finally inducing oxidative stress in myotubes. Consistently, serine dietary deprivation strongly strengthens cancer-associated weight loss and muscle atrophy in mice models. These findings uncover serine consumption by tumor cells as a previously undisclosed driver in cancer cachexia, opening new routes for possible therapeutic approaches.
Collapse
Affiliation(s)
- Erica Pranzini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Livio Muccillo
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
- AREA Science Park, Padriciano, 99, Trieste, Italy
| | - Ilaria Nesi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Alice Santi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Caterina Mancini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Giulia Lori
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Massimo Genovese
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Tiziano Lottini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giuseppina Comito
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Anna Caselli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lina Sabatino
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Vittorio Colantuoni
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Maria Letizia Taddei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Paolo Cirri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Paolo Paoli
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| |
Collapse
|
3
|
Yu J, Spielvogel C, Haberl D, Jiang Z, Özer Ö, Pusitz S, Geist B, Beyerlein M, Tibu I, Yildiz E, Kandathil SA, Buschhorn T, Schnöll J, Kumpf K, Chen YT, Wu T, Zhang Z, Grünert S, Hacker M, Vraka C. Systemic Metabolic and Volumetric Assessment via Whole-Body [ 18F]FDG-PET/CT: Pancreas Size Predicts Cachexia in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2024; 16:3352. [PMID: 39409971 PMCID: PMC11475137 DOI: 10.3390/cancers16193352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/27/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objectives: Cancer-associated cachexia in head and neck squamous cell carcinoma (HNSCC) is challenging to diagnose due to its complex pathophysiology. This study aimed to identify metabolic biomarkers linked to cachexia and survival in HNSCC patients using [18F]FDG-PET/CT imaging and machine learning (ML) techniques. Methods: We retrospectively analyzed 253 HNSCC patients from Vienna General Hospital and the MD Anderson Cancer Center. Automated organ segmentation was employed to quantify metabolic and volumetric data from [18F]FDG-PET/CT scans across 29 tissues and organs. Patients were categorized into low weight loss (LoWL; grades 0-2) and high weight loss (HiWL; grades 3-4) groups, according to the weight loss grading system (WLGS). Machine learning models, combined with Cox regression, were used to identify survival predictors. Shapley additive explanation (SHAP) analysis was conducted to determine the significance of individual features. Results: The HiWL group exhibited increased glucose metabolism in skeletal muscle and adipose tissue (p = 0.01), while the LoWL group showed higher lung metabolism. The one-year survival rate was 84.1% in the LoWL group compared to 69.2% in the HiWL group (p < 0.01). Pancreatic volume emerged as a key biomarker associated with cachexia, with the ML model achieving an AUC of 0.79 (95% CI: 0.77-0.80) and an accuracy of 0.82 (95% CI: 0.81-0.83). Multivariate Cox regression confirmed pancreatic volume as an independent prognostic factor (HR: 0.66, 95% CI: 0.46-0.95; p < 0.05). Conclusions: The integration of metabolic and volumetric data provided a strong predictive model, highlighting pancreatic volume as a key imaging biomarker in the metabolic assessment of cachexia in HNSCC. This finding enhances our understanding and may improve prognostic evaluations and therapeutic strategies.
Collapse
Affiliation(s)
- Josef Yu
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Clemens Spielvogel
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - David Haberl
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, 1090 Vienna, Austria
| | - Zewen Jiang
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Öykü Özer
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Smilla Pusitz
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Barbara Geist
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Michael Beyerlein
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Iustin Tibu
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Erdem Yildiz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, 1090 Vienna, Austria; (E.Y.); (S.A.K.); (T.B.); (J.S.)
| | - Sam Augustine Kandathil
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, 1090 Vienna, Austria; (E.Y.); (S.A.K.); (T.B.); (J.S.)
| | - Till Buschhorn
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, 1090 Vienna, Austria; (E.Y.); (S.A.K.); (T.B.); (J.S.)
| | - Julia Schnöll
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Vienna, 1090 Vienna, Austria; (E.Y.); (S.A.K.); (T.B.); (J.S.)
| | - Katarina Kumpf
- IT4Science, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ying-Ting Chen
- Teaching Center, Medical University of Vienna, 1090 Vienna, Austria;
| | - Tingting Wu
- Department of Cardiology, Xiangya Hospital Central South University, Changsha 410008, China;
| | - Zhaoqi Zhang
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050010, China;
| | - Stefan Grünert
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| | - Chrysoula Vraka
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria; (J.Y.); (C.S.); (D.H.); (Z.J.); (Ö.Ö.); (S.P.); (B.G.); (S.G.); (M.H.)
| |
Collapse
|
4
|
Lan XQ, Deng CJ, Wang QQ, Zhao LM, Jiao BW, Xiang Y. The role of TGF-β signaling in muscle atrophy, sarcopenia and cancer cachexia. Gen Comp Endocrinol 2024; 353:114513. [PMID: 38604437 DOI: 10.1016/j.ygcen.2024.114513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/24/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
Skeletal muscle, comprising a significant proportion (40 to 50 percent) of total body weight in humans, plays a critical role in maintaining normal physiological conditions. Muscle atrophy occurs when the rate of protein degradation exceeds protein synthesis. Sarcopenia refers to age-related muscle atrophy, while cachexia represents a more complex form of muscle wasting associated with various diseases such as cancer, heart failure, and AIDS. Recent research has highlighted the involvement of signaling pathways, including IGF1-Akt-mTOR, MuRF1-MAFbx, and FOXO, in regulating the delicate balance between muscle protein synthesis and breakdown. Myostatin, a member of the TGF-β superfamily, negatively regulates muscle growth and promotes muscle atrophy by activating Smad2 and Smad3. It also interacts with other signaling pathways in cachexia and sarcopenia. Inhibition of myostatin has emerged as a promising therapeutic approach for sarcopenia and cachexia. Additionally, other TGF-β family members, such as TGF-β1, activin A, and GDF11, have been implicated in the regulation of skeletal muscle mass. Furthermore, myostatin cooperates with these family members to impair muscle differentiation and contribute to muscle loss. This review provides an overview of the significance of myostatin and other TGF-β signaling pathway members in muscular dystrophy, sarcopenia, and cachexia. It also discusses potential novel therapeutic strategies targeting myostatin and TGF-β signaling for the treatment of muscle atrophy.
Collapse
Affiliation(s)
- Xin-Qiang Lan
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Cheng-Jie Deng
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Qi-Quan Wang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Li-Min Zhao
- Senescence and Cancer Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China
| | - Bao-Wei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, Yunnan, China
| | - Yang Xiang
- Metabolic Control and Aging Group, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, Nanchang 330031, Jiangxi, China.
| |
Collapse
|
5
|
Lara O, Janssen P, Mambretti M, De Pauw L, Ates G, Mackens L, De Munck J, Walckiers J, Pan Z, Beckers P, Espinet E, Sato H, De Ridder M, Marks DL, Barbé K, Aerts JL, Hermans E, Rooman I, Massie A. Compartmentalized role of xCT in supporting pancreatic tumor growth, inflammation and mood disturbance in mice. Brain Behav Immun 2024; 118:275-286. [PMID: 38447884 DOI: 10.1016/j.bbi.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/05/2024] [Accepted: 03/02/2024] [Indexed: 03/08/2024] Open
Abstract
xCT (Slc7a11), the specific subunit of the cystine/glutamate antiporter system xc-, is present in the brain and on immune cells, where it is known to modulate behavior and inflammatory responses. In a variety of cancers -including pancreatic ductal adenocarcinoma (PDAC)-, xCT is upregulated by tumor cells to support their growth and spread. Therefore, we studied the impact of xCT deletion in pancreatic tumor cells (Panc02) and/or the host (xCT-/- mice) on tumor burden, inflammation, cachexia and mood disturbances. Deletion of xCT in the tumor strongly reduced tumor growth. Targeting xCT in the host and not the tumor resulted only in a partial reduction of tumor burden, while it did attenuate tumor-related systemic inflammation and prevented an increase in immunosuppressive regulatory T cells. The latter effect could be replicated by specific xCT deletion in immune cells. xCT deletion in the host or the tumor differentially modulated neuroinflammation. When mice were grafted with xCT-deleted tumor cells, hypothalamic inflammation was reduced and, accordingly, food intake improved. Tumor bearing xCT-/- mice showed a trend of reduced hippocampal neuroinflammation with less anxiety- and depressive-like behavior. Taken together, targeting xCT may have beneficial effects on pancreatic cancer-related comorbidities, beyond reducing tumor burden. The search for novel and specific xCT inhibitors is warranted as they may represent a holistic therapy in pancreatic cancer.
Collapse
Affiliation(s)
- Olaya Lara
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Janssen
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium; Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Marco Mambretti
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Laura De Pauw
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Gamze Ates
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Liselotte Mackens
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jolien De Munck
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Jarne Walckiers
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Zhaolong Pan
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium
| | - Pauline Beckers
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Elisa Espinet
- Pancreatic Cancer Lab, Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona, L'Hospitalet de Llobregat, Barcelona 08907, Spain; Molecular Mechanisms and Experimental Therapy in Oncology Program, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Hideyo Sato
- Department of Medical Technology, Niigata University, Niigata 950-3198, Japan
| | - Mark De Ridder
- Department of Radiotherapy, UZ Brussels, VUB, Brussels 1090, Belgium
| | - Daniel L Marks
- Papé Family Pediatric Research Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kurt Barbé
- The Biostatistics and Medical Informatics Department, VUB, Brussels 1090, Belgium
| | - Joeri L Aerts
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium
| | - Emmanuel Hermans
- Institute of Neuroscience, Université catholique de Louvain, Brussels 1200, Belgium
| | - Ilse Rooman
- Laboratory for Medical and Molecular Oncology, Translational Oncology Research Center (TORC), VUB, Brussels 1090, Belgium.
| | - Ann Massie
- Laboratory of Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences (C4N), Vrije Universiteit Brussel (VUB), Brussels 1090, Belgium.
| |
Collapse
|
6
|
Chung MH, Chang WP. Correlation between hemoglobin levels and depression in late-stage cancer patients with irritability as mediating variable. Eur J Oncol Nurs 2023; 67:102414. [PMID: 37804750 DOI: 10.1016/j.ejon.2023.102414] [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: 06/04/2023] [Revised: 07/30/2023] [Accepted: 09/07/2023] [Indexed: 10/09/2023]
Abstract
PURPOSE In late-stage cancer, the cancer itself or the side effects of cancer treatment are known to affect the hemoglobin (Hgb) levels or emotions of patients. We to investigate the relationship between Hgb levels and depression in late-stage cancer patients and verified whether irritability has a mediating effect on this relationship. METHOD The research tools included a patient basic information form, the Irritability Scale-Initial Version (TISi), and the Hamilton Depression Rating Scale (HAMD). We first compared the Hgb levels, HAMD scores, and TISi scores of the cancer patients with different attributes, performed multiple hierarchical regression analysis, and then analyzed the mediating effects of TISi scores using the Sobel test. RESULTS In the 117 late-stage cancer patients, Hgb levels of patients with a BMI<18.5 kg/m2 were lower than those of the patients with a BMI 24.0 kg/m2. Hgb levels had a negative influence on both TISi scores (B = -2.74, p = .001) and HAMD scores (B = -0.75, p = .010). TISi scores mediated the relationship between Hgb levels and HAMD scores (Z = 2.06, p = .040). CONCLUSIONS Irritability is a mediating variable of the influence of Hgb levels on depression, meaning that lower Hgb levels in late-stage cancer patients may be detrimental to emotional stability, induce irritability, and thereby cause depression. Thus, in the psychological care of late-stage cancer patients, medical teams should be more vigilant in monitoring Hgb levels and anemia treatment.
Collapse
Affiliation(s)
- Min-Huey Chung
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan
| | - Wen-Pei Chang
- School of Nursing, College of Nursing, Taipei Medical University, Taipei, Taiwan; Department of Nursing, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
7
|
Shivnani P, Shekhawat S, Prajapati A. Cancer Cachexia and breast cancer stem cell signalling - A crosstalk of signalling molecules. Cell Signal 2023; 110:110847. [PMID: 37557973 DOI: 10.1016/j.cellsig.2023.110847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/21/2023] [Accepted: 08/05/2023] [Indexed: 08/11/2023]
Abstract
Cancer Cachexia is a condition characterized by the involuntary loss of lean body mass, a negative protein and energy balance, and systemic inflammation. This syndrome profoundly impacts the patient's quality of life and is linked to poor chemotherapy response and reduced survival. Despite multiple mechanisms being implicated in its development, and various cytokines believed to contribute to the persistent catabolic state, cachexia is still not fully recognized and is often left untreated. Cachexia is caused by altered metabolic adaptation and lack of anticactic therapy due to systemic cytokines promoting and fuelling cancer growth. The exact molecular mechanisms and clinical endpoints remain poorly defined. It has an occurrence rate of 30%-80%, accounting for 20% of total cancer mortality. Tumor cells remodel the microenvironment suitable for their proliferation, wherein they communicate with fibroblast cells to modulate their expression and induce tumor progressive cytokines. Several studies have reported its strong correlation with systemic cytokines that initiate and aggravate the condition. Plenty of studies show the prominent role of cancer-induced cachexia in pancreatic cancer, colon cancer, and lung cancer. However, limited data are available for breast cancer-induced cachexia, highlighting the need for studying it. Breast cancer stem cells (BCSCs) are a prominently explored area in breast cancer research. They are characterized by CD44+/CD24-/ALDH+ expression and are a focus of cancer research. They are a source of renewal and differentiation within the tumor environment and are responsible for progression, and chemotherapeutic resistance. The tumor microenvironment and its cytokines are responsible for maintaining and inducing their differentiation. Cytokines significantly impact BCSC development and self-renewal, stimulating or inhibiting proliferation depending on cytokine and environment. Pro-inflammatory mediators like IL-6, TNF-α, and IL-8 increase proliferation, promoting tumor growth. Experimental models and clinical studies have shown a direct relationship between cytokines and BCSC proliferation. Several of them seem to be interconnected as they initiate signalling down different pathways but converge at BCSC increase and tumor proliferation. This review highlights the common pathways between cachexia and BCSC signalling, to identify potential therapeutic targets that can aid both conditions.
Collapse
Affiliation(s)
- Priyanka Shivnani
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India
| | - Saroj Shekhawat
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India
| | - Akhilesh Prajapati
- Biotechnology, School of Science, GSFC University, Vadodara 391750, India.
| |
Collapse
|
8
|
Agulló-Ortuño MT, Mancebo E, Grau M, Núñez Sobrino JA, Paz-Ares L, López-Martín JA, Flández M. Tryptophan Modulation in Cancer-Associated Cachexia Mouse Models. Int J Mol Sci 2023; 24:13005. [PMID: 37629186 PMCID: PMC10455959 DOI: 10.3390/ijms241613005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer cachexia is a multifactorial syndrome that interferes with treatment and reduces the quality of life and survival of patients. Currently, there is no effective treatment or biomarkers, and pathophysiology is not clear. Our group reported alterations on tryptophan metabolites in cachectic patients, so we aim to investigate the role of tryptophan using two cancer-associated cachexia syngeneic murine models, melanoma B16F10, and pancreatic adenocarcinoma that is KPC-based. Injected mice showed signs of cancer-associated cachexia as reduction in body weight and raised spleen weight, MCP1, and carbonilated proteins in plasma. CRP and Myostatin also increased in B16F10 mice. Skeletal muscle showed a decrease in quadriceps weight and cross-sectional area (especially in B16F10). Higher expression of atrophy genes, mainly Atrogin1, was also observed. Plasmatic tryptophan levels in B16F10 tumor-bearing mice decreased even at early steps of tumorigenesis. In KPC-injected mice, tryptophan fluctuated but were also reduced and in cachectic patients were significantly lower. Treatment with 1-methyl-tryptophan, an inhibitor of tryptophan degradation, in the murine models resulted in the restoration of plasmatic tryptophan levels and an improvement on splenomegaly and carbonilated proteins levels, while changes in plasmatic inflammatory markers were mild. After the treatment, CCR2 expression in monocytes diminished and lymphocytes, Tregs, and CD8+, were activated (seen by increased in CD127 and CD25 expression, respectively). These immune cell changes pointed to an improvement in systemic inflammation. While treatment with 1-MT did not show benefits in terms of muscle wasting and atrophy in our experimental setting, muscle functionality was not affected and central nuclei fibers appeared, being a feature of regeneration. Therefore, tryptophan metabolism pathway is a promising target for inflammation modulation in cancer-associated cachexia.
Collapse
Affiliation(s)
- M. Teresa Agulló-Ortuño
- Laboratory of Clinical and Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Av. Córdoba s/n, 28041 Madrid, Spain; (M.T.A.-O.); (L.P.-A.)
- Lung Cancer Group, Clinical Research Program, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
- Biomedical Research Networking Centre on Oncology—CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nursing, Facultad de Fisioterapia y Enfermería, Universidad de Castilla La-Mancha (UCLM), 45071 Toledo, Spain
| | - Esther Mancebo
- Department of Immunology, Hospital Universitario 12 de Octubre, Av. Córdoba s/n, 28041 Madrid, Spain;
| | - Montserrat Grau
- Animal Facility, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Av. Córdoba s/n, 28041 Madrid, Spain;
| | - Juan Antonio Núñez Sobrino
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Av. Córdoba s/n, 28041 Madrid, Spain;
| | - Luis Paz-Ares
- Laboratory of Clinical and Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Av. Córdoba s/n, 28041 Madrid, Spain; (M.T.A.-O.); (L.P.-A.)
- Lung Cancer Group, Clinical Research Program, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
- Biomedical Research Networking Centre on Oncology—CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Av. Córdoba s/n, 28041 Madrid, Spain;
- Medicine Department, Facultad de Medicina y Cirugía, Universidad Complutense de Madrid (UCM), 28040 Madrid, Spain
| | - José A. López-Martín
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Av. Córdoba s/n, 28041 Madrid, Spain;
| | - Marta Flández
- Laboratory of Clinical and Translational Oncology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), Av. Córdoba s/n, 28041 Madrid, Spain; (M.T.A.-O.); (L.P.-A.)
- Faculty of Experimental Sciences, Francisco de Vitoria University (UFV), 28223 Pozuelo de Alarcón, Spain
| |
Collapse
|
9
|
Argilés JM, López-Soriano FJ, Stemmler B, Busquets S. Cancer-associated cachexia - understanding the tumour macroenvironment and microenvironment to improve management. Nat Rev Clin Oncol 2023; 20:250-264. [PMID: 36806788 DOI: 10.1038/s41571-023-00734-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/22/2023]
Abstract
Cachexia is a devastating, multifactorial and often irreversible systemic syndrome characterized by substantial weight loss (mainly of skeletal muscle and adipose tissue) that occurs in around 50-80% of patients with cancer. Although this condition mainly affects skeletal muscle (which accounts for approximately 40% of total body weight), cachexia is a multi-organ syndrome that also involves white and brown adipose tissue, and organs including the bones, brain, liver, gut and heart. Notably, cachexia accounts for up to 20% of cancer-related deaths. Cancer-associated cachexia is invariably associated with systemic inflammation, anorexia and increased energy expenditure. Understanding these mechanisms is essential, and the progress achieved in this area over the past decade could help to develop new therapeutic approaches. In this Review, we examine the currently available evidence on the roles of both the tumour macroenvironment and microenvironment in cancer-associated cachexia, and provide an overview of the novel therapeutic strategies developed to manage this syndrome.
Collapse
Affiliation(s)
- Josep M Argilés
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain.
| | - Francisco J López-Soriano
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| | | | - Silvia Busquets
- Cancer Research Group, Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
- Institut de Biomedicina de la Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
10
|
In vitro chemotherapy-associated muscle toxicity is attenuated with nutritional support, while treatment efficacy is retained. Oncotarget 2022; 13:1094-1108. [PMID: 36242541 PMCID: PMC9564364 DOI: 10.18632/oncotarget.28279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Purpose: Muscle-wasting and treatment-related toxicities negatively impact prognosis of colorectal cancer (CRC) patients. Specific nutritional composition might support skeletal muscle and enhance treatment support. In this in vitro study we assess the effect of nutrients EPA, DHA, L-leucine and vitamin D3, as single nutrients or in combination on chemotherapy-treated C2C12-myotubes, and specific CRC-tumor cells. Materials and Methods: Using C2C12-myotubes, the effects of chemotherapy (oxaliplatin, 5-fluorouracil, oxaliplatin+5-fluorouracil and irinotecan) on protein synthesis, cell-viability, caspase-3/7-activity and LDH-activity were assessed. Addition of EPA, DHA, L-leucine and vitamin D3 and their combination (SNCi) were studied in presence of above chemotherapies. Tumor cell-viability was assessed in oxaliplatin-treated C26 and MC38 CRC cells, and in murine and patient-derived CRC-organoids. Results: While chemotherapy treatment of C2C12-myotubes decreased protein synthesis, cell-viability and increased caspase-3/7 and LDH-activity, SNCi showed improved protein synthesis and cell viability and lowered LDH activity. The nutrient combination SNCi showed a better overall performance compared to the single nutrients. Treatment response of tumor models was not significantly affected by addition of nutrients. Conclusions: This in vitro study shows protective effect with specific nutrition composition of C2C12-myotubes against chemotherapy toxicity, which is superior to the single nutrients, while treatment response of tumor cells remained.
Collapse
|
11
|
Shin E, Kang H, Lee H, Lee S, Jeon J, Seong K, Youn H, Youn B. Exosomal Plasminogen Activator Inhibitor-1 Induces Ionizing Radiation-Adaptive Glioblastoma Cachexia. Cells 2022; 11:cells11193102. [PMID: 36231065 PMCID: PMC9564109 DOI: 10.3390/cells11193102] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/14/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022] Open
Abstract
Cancer cachexia is a muscle-wasting syndrome that leads to a severely compromised quality of life and increased mortality. A strong association between cachexia and poor prognosis has been demonstrated in intractable cancers, including glioblastoma (GBM). In the present study, it was demonstrated that ionizing radiation (IR), the first-line treatment for GBM, causes cancer cachexia by increasing the exosomal release of plasminogen activator inhibitor-1 (PAI-1) from glioblastoma cells. Exosomal PAI-1 delivered to the skeletal muscle is directly penetrated in the muscles and phosphorylates STAT3 to intensify muscle atrophy by activating muscle RING-finger protein-1 (MuRF1) and muscle atrophy F-box (Atrogin1); furthermore, it hampers muscle protein synthesis by inhibiting mTOR signaling. Additionally, pharmacological inhibition of PAI-1 by TM5441 inhibited muscle atrophy and rescued muscle protein synthesis, thereby providing survival benefits in a GBM orthotopic xenograft mouse model. In summary, our data delineated the role of PAI-1 in the induction of GBM cachexia associated with radiotherapy-treated GBM. Our data also indicated that targeting PAI-1 could serve as an attractive strategy for the management of GBM following radiotherapy, which would lead to a considerable improvement in the quality of life of GBM patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Eunguk Shin
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Haksoo Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
| | - Jaewan Jeon
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University College of Medicine, Busan 48108, Korea
| | - Kimoon Seong
- Laboratory of Biological Dosimetry, National Radiation Emergency Medical Center (NREMC), Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul 01812, Korea
| | - Hyesook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul 05006, Korea
| | - Buhyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan 46241, Korea
- Department of Biological Sciences, Pusan National University, Busan 46241, Korea
- Correspondence: ; Tel.: +82-51-510-2264
| |
Collapse
|
12
|
Adipose Tissue Wasting as a Determinant of Pancreatic Cancer-Related Cachexia. Cancers (Basel) 2022; 14:cancers14194754. [PMID: 36230682 PMCID: PMC9563866 DOI: 10.3390/cancers14194754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Pancreatic cancer (PC) is one of the deadliest cancers in the US. The poor prognosis of PC is related to diagnostic delay and the presence of unintended weight loss (cachexia) that commonly presents in PC patients even before diagnosis. However, the current understanding of how PC mediates cachexia is limited, and there are few treatments clinically available for cachexia. Based on the current literature, we demonstrate that PC-related cachexia primarily results from the wasting of adipose tissue, once thought to be merely a storage depot but now appreciated as an instrumental metabolic organ in the body. In addition, poor survival in PC patients was found to be associated with adipose tissue loss at diagnosis and during treatment. Therefore, identifying potential mediators and molecular mechanisms underlying adipose tissue loss would promise to pave the way for the development of effective interventions for PC-related cachexia Abstract Pancreatic cancer (PC) is the third leading cause of cancer-related death in the US, and its 5-year survival rate is approximately 10%. The low survival rates largely stem from diagnostic delay and the presence of significant adipose tissue and muscle wasting, commonly referred to as cachexia. Cachexia is present in nearly 80% of PC patients and is a key cause of poor response to treatment and about 20% of death in PC patients. However, there are few clinical interventions proven to be effective against PC-related cachexia. Different cancer types feature distinct secretome profiles and functional characteristics which would lead to cachexia development differently. Therefore, here we discuss affected tissues and potential mechanisms leading to cachexia in PC. We postulate that the most affected tissue during the development of PC-related cachexia is adipose tissue, historically and still thought to be just an inert repository for excess energy in relation to cancer-related cachexia. Adipose tissue loss is considerably greater than muscle loss in quantity and shows a correlation with poor survival in PC patients. Moreover, we suggest that PC mediates adipose atrophy by accelerating adipocyte lipid turnover and fibroblast infiltration.
Collapse
|
13
|
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: 16] [Impact Index Per Article: 5.3] [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
|
14
|
Cancer- and cardiac-induced cachexia: same fate through different inflammatory mediators? Inflamm Res 2022; 71:771-783. [PMID: 35680678 DOI: 10.1007/s00011-022-01586-y] [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: 11/17/2021] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Inflammation is widely recognized as the driving force of cachexia induced by chronic diseases; however, therapies targeting inflammation do not always reverse cachexia. Thus, whether inflammation per se plays an important role in the clinical course of cachectic patients is still a matter of debate. AIMS To give new insights into cachexia's pathogenesis and diagnosis, we performed a comprehensive literature search on the contribution of inflammatory markers to this syndrome, focusing on the noncommunicable diseases cancer and cardiovascular diseases. METHODS A systematic review was performed in PubMed using the keywords ("cancer" OR "cardiac" cachexia AND "human" OR "patient" AND "plasma" or "serum"). A total of 744 studies were retrieved and, from these, 206 were selected for full-text screening. In the end, 98 papers focusing on circulating biomarkers of cachexia were identified, which resulted in a list of 113 different mediators. RESULTS Data collected from the literature highlight the contribution of interleukin-6 (IL-6) and C-reactive protein (CRP) to cachexia, independently of the underlying condition. Despite not being specific, once the diagnosis of cachexia is established, CRP might help to monitor the effectiveness of anti-cachexia therapies. In cardiac diseases, B-type natriuretic peptide (BNP), renin, and obestatin might be putative markers of body wasting, whereas in cancer, growth differentiation factor (GDF) 15, transforming growth factor (TGF)-β1 and vascular endothelial growth factor (VEGF) C seem to be better markers of this syndrome. Independently of the circulating mediators, NF-κB and JAK/STAT signaling pathways play a key role in bridging inflammation with muscle wasting; however, therapies targeting these pathways were not proven effective for all cachectic patients. CONCLUSION The critical and integrative analysis performed herein will certainly feed future research focused on the better comprehension of cachexia pathogenesis toward the improvement of its diagnosis and the development of personalized therapies targeting specific cachexia phenotypes.
Collapse
|
15
|
Celichowska M, Miedziaszczyk M, Lacka K. Pharmacotherapy in Cachexia: A Review of Endocrine Abnormalities and Steroid Pharmacotherapy. J Pain Palliat Care Pharmacother 2022; 36:117-131. [PMID: 35758863 DOI: 10.1080/15360288.2022.2063469] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Cachexia is a state of increased metabolism associated with high morbidity and mortality. Dysregulation of cytokines and hormone activity causes reduced protein synthesis and excessive protein breakdown. various treatments are available, depending on the primary disease and the patient's state. Besides pharmacological treatment, crucial is nutritional support as well as increasing physical activity. The main purpose of pharmacological treatment is to diminish inflammation, improve appetite and decrease muscle wasting. Therefore a lot of medications aim at proinflammatory cytokines such as Interferon-α or Tumor Necrosis Factor-β, but because of the complicated mechanism of cachexia, the range of targets is very wide. in cachexia treatment, use of corticosteroids is common, which improve appetite, diminish inflammation, inhibit prostaglandin metabolism, Interleukin-1 activity. They can also decrease protein synthesis and increase protein degradation, which can be prevented by resveratrol. Estrogen analogs, progesterone analogs, testosterone analogs, Selective Androgen Receptor Modulators (SARM), Angiotensin-Converting-Enzyme Inhibitors (ACEI), Nonsteroidal anti-inflammatory drugs (NSAIDs), thalidomide, melatonin, Growth Hormone Releasing Peptide-2 (GHRP-2) may play important role in wasting syndrome treatment as well. However, for the usage of some of them, evidence-based recommendations are not available. This review highlights current therapeutic options for cachexia with a specific focus on steroid therapy.
Collapse
Affiliation(s)
- Magdalena Celichowska
- Magdalena Celichowska is with the Student's Scientific Group of Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Miłosz Miedziaszczyk
- Miłosz Miedziaszczyk is with the Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Katarzyna Lacka
- Katarzyna Lacka,MD, PhD is with the Department of Endocrinology, Metabolism and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
16
|
Exercise Counteracts the Deleterious Effects of Cancer Cachexia. Cancers (Basel) 2022; 14:cancers14102512. [PMID: 35626116 PMCID: PMC9139714 DOI: 10.3390/cancers14102512] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary This review provides an overview of the effects of exercise training on the major mechanisms related to cancer cachexia (CC). The review also discusses how cancer comorbidities can influence the ability of patients/animals with cancer to perform exercise training and what precautions should be taken when they exercise. The contribution of other factors, such as exercise modality and biological sex, to exercise effectiveness in ameliorating CC are also elaborated in the final sections. We provide meticulous evidence for how advantageous exercise training can be in patients/animals with CC at molecular and cellular levels. Finally, we emphasise what factors should be considered to optimise and personalise an exercise training program in CC. Abstract Cancer cachexia (CC) is a multifactorial syndrome characterised by unintentional loss of body weight and muscle mass in patients with cancer. The major hallmarks associated with CC development and progression include imbalanced protein turnover, inflammatory signalling, mitochondrial dysfunction and satellite cell dysregulation. So far, there is no effective treatment to counteract muscle wasting in patients with CC. Exercise training has been proposed as a potential therapeutic approach for CC. This review provides an overview of the effects of exercise training in CC-related mechanisms as well as how factors such as cancer comorbidities, exercise modality and biological sex can influence exercise effectiveness in CC. Evidence in mice and humans suggests exercise training combats all of the hallmarks of CC. Several exercise modalities induce beneficial adaptations in patients/animals with CC, but concurrent resistance and endurance training is considered the optimal type of exercise. In the case of cancer patients presenting comorbidities, exercise training should be performed only under specific guidelines and precautions to avoid adverse effects. Observational comparison of studies in CC using different biological sex shows exercise-induced adaptations are similar between male and female patients/animals with cancer, but further studies are needed to confirm this.
Collapse
|
17
|
Silva D, Quintas C, Gonçalves J, Fresco P. Contribution of adrenergic mechanisms for the stress-induced breast cancer carcinogenesis. J Cell Physiol 2022; 237:2107-2127. [PMID: 35243626 DOI: 10.1002/jcp.30707] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 02/12/2022] [Indexed: 12/11/2022]
Abstract
Breast cancer is the most common and deadliest type of cancer in women. Stress exposure has been associated with carcinogenesis and the stress released neurotransmitters, noradrenaline and adrenaline, and their cognate receptors, can participate in the carcinogenesis process, either by regulating tumor microenvironment or by promoting systemic changes. This work intends to provide an overview of the research done in this area and try to unravel the role of adrenergic ligands in the context of breast carcinogenesis. In the initiation phase, adrenergic signaling may favor neoplastic transformation of breast epithelial cells whereas, during cancer progression, may favor the metastatic potential of breast cancer cells. Additionally, adrenergic signaling can alter the function and activity of other cells present in the tumor microenvironment towards a protumor phenotype, namely macrophages, fibroblasts, and by altering adipocyte's function. Adrenergic signaling also promotes angiogenesis and lymphangiogenesis and, systemically, may induce the formation of preneoplastic niches, cancer-associated cachexia and alterations in the immune system which contribute for the loss of quality of life of breast cancer patients and their capacity to fight cancer. Most studies points to a major contribution of β2 -adrenoceptor activated pathways on these effects. The current knowledge of the mechanistic pathways activated by β2 -adrenoceptors in physiology and pathophysiology, the availability of selective drugs approved for clinical use and a deeper knowledge of the basic cellular and molecular pathways by which adrenergic stimulation may influence cancer initiation and progression, opens the possibility to use new therapeutic alternatives to improve efficacy of breast cancer treatments.
Collapse
Affiliation(s)
- Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Clara Quintas
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Simon L, Baldwin C, Kalea AZ, Slee A. Cannabinoid interventions for improving cachexia outcomes in cancer: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle 2022; 13:23-41. [PMID: 34881518 PMCID: PMC8818598 DOI: 10.1002/jcsm.12861] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 01/06/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a wasting syndrome characterized by involuntary weight loss and anorexia. Clear definition and diagnostic criteria for CAC are lacking, which makes it difficult to estimate its prevalence, to interpret research and to compare studies. There is no standard treatment to manage CAC, but previous studies support the use of cannabinoids for cachexia in other chronic diseases including HIV and multiple sclerosis. However, only a few randomized controlled trials (RCTs) and one meta-analysis of this intervention in cancer populations are available. Non-randomized studies of interventions (NRSIs) are often excluded from reviews due to variable methodology and potential for biases. This review aimed to consider NRSIs alongside RCTs to provide a complete summary of the available evidence that clinical decision makers could use in future investigations. Literature searches were conducted using three databases for relevant RCTs or NRSIs according to Cochrane methodology. Abstract and full texts of retrieved manuscripts were selected and retrieved by two investigators based on the PRISMA-A guidelines, and risk of bias and quality of evidence assessments were performed. Outcome data on weight, appetite, quality of life, performance status, adverse effects, and mortality were combined by narrative synthesis and meta-analysis where possible. Ten studies were included, four of which were RCTs and six NRSIs matching the eligibility criteria. Very low-quality evidence from meta-analysis suggested no significant benefits of cannabinoids for appetite compared with control (standardized mean difference: -0.02; 95% confidence interval: -0.51, 0.46; P = 0.93). Patient-reported observations from NRSIs suggested improvements in appetite. Another meta-analysis of moderate quality evidence showed that cannabinoids were significantly less efficient than active or inactive control on quality of life (standardized mean difference: -0.25; 95% confidence interval: -0.43, -0.07; P = 0.007). The effectiveness of cannabinoids alone to improve outcomes of CAC remains unclear. Low-quality evidence from both RCTs and NRSIs shows no significant benefits of cannabinoids for weight gain, appetite stimulation, and better quality of life, three important outcomes of cachexia. Higher quality research integrating cannabinoids into multi-modal therapies may offer better opportunities for developing CAC-specific treatments. This review also highlights that findings from non-randomized studies of interventions (NRSIs) can provide evidence of the effects of an intervention and advocate for the feasibility of larger RCTs.
Collapse
Affiliation(s)
- Lucile Simon
- Division of MedicineUniversity College LondonLondonUK
| | | | - Anastasia Z. Kalea
- Division of MedicineUniversity College LondonLondonUK
- Institute of Cardiovascular SciencesUniversity College LondonLondonUK
| | - Adrian Slee
- Division of MedicineUniversity College LondonLondonUK
| |
Collapse
|
19
|
Towards Drug Repurposing in Cancer Cachexia: Potential Targets and Candidates. Pharmaceuticals (Basel) 2021; 14:ph14111084. [PMID: 34832866 PMCID: PMC8618795 DOI: 10.3390/ph14111084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/11/2022] Open
Abstract
As a multifactorial and multiorgan syndrome, cancer cachexia is associated with decreased tolerance to antitumor treatments and increased morbidity and mortality rates. The current approaches for the treatment of this syndrome are not always effective and well established. Drug repurposing or repositioning consists of the investigation of pharmacological components that are already available or in clinical trials for certain diseases and explores if they can be used for new indications. Its advantages comparing to de novo drugs development are the reduced amount of time spent and costs. In this paper, we selected drugs already available or in clinical trials for non-cachexia indications and that are related to the pathways and molecular components involved in the different phenotypes of cancer cachexia syndrome. Thus, we introduce known drugs as possible candidates for drug repurposing in the treatment of cancer-induced cachexia.
Collapse
|
20
|
Anti-Cancer Effects of Cyclic Peptide ALOS4 in a Human Melanoma Mouse Model. Int J Mol Sci 2021; 22:ijms22179579. [PMID: 34502483 PMCID: PMC8430629 DOI: 10.3390/ijms22179579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/17/2022] Open
Abstract
We examined the effects of ALOS4, a cyclic peptide discovered previously by phage library selection against integrin αvβ3, on a human melanoma (A375) xenograft model to determine its abilities as a potential anti-cancer agent. We found that ALOS4 promoted healthy weight gain in A375-engrafted nude mice and reduced melanoma tumor mass and volume. Despite these positive changes, examination of the tumor tissue did not indicate any significant effects on proliferation, mitotic index, tissue vascularization, or reduction of αSMA or Ki-67 tumor markers. Modulation in overall expression of critical downstream αvβ3 integrin factors, such as FAK and Src, as well as reductions in gene expression of c-Fos and c-Jun transcription factors, indirectly confirmed our suspicions that ALOS4 is likely acting through an integrin-mediated pathway. Further, we found no overt formulation issues with ALOS4 regarding interaction with standard inert laboratory materials (polypropylene, borosilicate glass) or with pH and temperature stability under prolonged storage. Collectively, ALOS4 appears to be safe, chemically stable, and produces anti-cancer effects in a human xenograft model of melanoma. We believe these results suggest a role for ALOS4 in an integrin-mediated pathway in exerting its anti-cancer effects possibly through immune response modulation.
Collapse
|
21
|
Kinoshita F, Tagawa T, Yamashita T, Takenaka T, Matsubara T, Toyokawa G, Takada K, Oba T, Osoegawa A, Yamazaki K, Takenoyama M, Shimokawa M, Nakashima N, Mori M. Prognostic value of postoperative decrease in serum albumin on surgically resected early-stage non-small cell lung carcinoma: A multicenter retrospective study. PLoS One 2021; 16:e0256894. [PMID: 34473762 PMCID: PMC8412276 DOI: 10.1371/journal.pone.0256894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/18/2021] [Indexed: 01/27/2023] Open
Abstract
Background Preoperative nutritional status is an important host-related prognostic factor for non-small cell lung carcinoma (NSCLC); however, the significance of postoperative changes in nutritional status remains unclear. This study aimed to elucidate the significance of postoperative decreases in serum albumin (ΔAlb) on the outcomes of early-stage NSCLC. Methods We analyzed 443 training cohort (TC) and 642 validation cohort (VC) patients with pStage IA NSCLC who underwent surgery and did not recur within 1 year. We measured preoperative serum albumin levels (preAlb) and postoperative levels 1 year after surgery (postAlb), and calculated ΔAlb as (preAlb − postAlb)/preAlb × 100%. A cutoff value of 11% for ΔAlb was defined on the basis of the receiver operating characteristic curve for the TC. Results Patients were divided into ΔAlb-Decreased and ΔAlb-Stable groups, including 100 (22.6%) and 343 (77.4%) in the TC, and 58 (9.0%) and 584 (90.1%) in the VC. ΔAlb-Decreased was associated with male sex (p = 0.0490), smoking (p = 0.0156), and non-adenocarcinoma (p<0.0001) in the TC, and pT1b (p = 0.0169) and non-adenocarcinoma (p = 0.0251) in the VC. Multivariable analysis identified ΔAlb as an independent prognostic factor for disease-free survival (DFS) and overall survival (OS) in both cohorts (VC: DFS, HR = 1.9, 95%CI: 1.10–3.15, p = 0.0197; OS, HR = 2.0, 95%CI: 1.13–3.45, p = 0.0173). Moreover, subgroup analysis demonstrated that the prognostic value of ΔAlb was consistent for age, sex, smoking history, surgical procedure, and histological type. Conclusion We demonstrated a negative impact of postoperative decrease of the serum albumin on the prognosis of patients with early-stage NSCLC. Postoperative changes in nutritional status might be important in NSCLC outcomes.
Collapse
Affiliation(s)
- Fumihiko Kinoshita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuzo Tagawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- * E-mail:
| | | | - Tomoyoshi Takenaka
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taichi Matsubara
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Gouji Toyokawa
- Department of Thoracic Surgery, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan
| | - Kazuki Takada
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Taro Oba
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Osoegawa
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koji Yamazaki
- Department of Thoracic Surgery, Clinical Research Institute, National Hospital Organization, Kyushu Medical Center, Fukuoka, Japan
| | - Mitsuhiro Takenoyama
- Department of Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Naoki Nakashima
- Medical Information Center, Kyushu University Hospital, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Moreira-Pais A, Ferreira R, Oliveira PA, Duarte JA. Sarcopenia versus cancer cachexia: the muscle wasting continuum in healthy and diseased aging. Biogerontology 2021; 22:459-477. [PMID: 34324116 DOI: 10.1007/s10522-021-09932-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022]
Abstract
Muscle wasting is one of the major health problems in older adults and is traditionally associated to sarcopenia. Nonetheless, muscle loss may also occur in older adults in the presence of cancer, and in this case, it is associated to cancer cachexia. The clinical management of these conditions is a challenge due to, at least in part, the difficulties in their differential diagnosis. Thus, efforts have been made to better comprehend the pathogenesis of sarcopenia and cancer cachexia, envisioning the improvement of their clinical discrimination and treatment. To add insights on this topic, this review discusses the current knowledge on key molecular players underlying sarcopenia and cancer cachexia in a comparative perspective. Data retrieved from this analysis highlight that while sarcopenia is characterized by the atrophy of fast-twitch muscle fibers, in cancer cachexia an increase in the proportion of fast-twitch fibers appears to happen. The molecular drivers for these specificmuscle remodeling patterns are still unknown; however, among the predominant contributors to sarcopenia is the age-induced neuromuscular denervation, and in cancer cachexia, the muscle disuse experienced by cancer patients seems to play an important role. Moreover, inflammation appears to be more severe in cancer cachexia. Impairment of nutrition-related mediators may also contribute to sarcopenia and cancer cachexia, being distinctly modulated in each condition.
Collapse
Affiliation(s)
- Alexandra Moreira-Pais
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal. .,LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal. .,Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal. .,Departamento de Química, Universidade de Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Paula A Oliveira
- Centre for Research and Technology of Agro Environmental and Biological Sciences (CITAB), Inov4Agro, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5000-801, Vila Real, Portugal
| | - José A Duarte
- CIAFEL, Faculty of Sport, University of Porto, Dr. Plácido da Costa 91, 4200-450, Porto, Portugal. .,Faculdade de Desporto, Universidade do Porto, Rua Dr. Plácido da Costa 91, 4200-450, Porto, Portugal. .,TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal.
| |
Collapse
|
23
|
Siff T, Parajuli P, Razzaque MS, Atfi A. Cancer-Mediated Muscle Cachexia: Etiology and Clinical Management. Trends Endocrinol Metab 2021; 32:382-402. [PMID: 33888422 PMCID: PMC8102392 DOI: 10.1016/j.tem.2021.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/12/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022]
Abstract
Muscle cachexia has a major detrimental impact on cancer patients, being responsible for 30% of all cancer deaths. It is characterized by a debilitating loss in muscle mass and function, which ultimately deteriorates patients' quality of life and dampens therapeutic treatment efficacy. Muscle cachexia stems from widespread alterations in whole-body metabolism as well as immunity and neuroendocrine functions and these global defects often culminate in aberrant signaling within skeletal muscle, causing muscle protein breakdown and attendant muscle atrophy. This review summarizes recent landmark discoveries that significantly enhance our understanding of the molecular etiology of cancer-driven muscle cachexia and further discuss emerging therapeutic approaches seeking to simultaneously target those newly discovered mechanisms to efficiently curb this lethal syndrome.
Collapse
Affiliation(s)
- Thomas Siff
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Parash Parajuli
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Mohammed S Razzaque
- Department of Pathology, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Azeddine Atfi
- Cellular and Molecular Pathogenesis Division, Department of Pathology and Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA; Sorbonne Universités, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France.
| |
Collapse
|
24
|
Cancer cachexia: molecular mechanism and pharmacological management. Biochem J 2021; 478:1663-1688. [PMID: 33970218 DOI: 10.1042/bcj20201009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022]
Abstract
Cancer cachexia often occurs in malignant tumors and is a multifactorial and complex symptom characterized by wasting of skeletal muscle and adipose tissue, resulting in weight loss, poor life quality and shorter survival. The pathogenic mechanism of cancer cachexia is complex, involving a variety of molecular substrates and signal pathways. Advancements in understanding the molecular mechanisms of cancer cachexia have provided a platform for the development of new targeted therapies. Although recent outcomes of early-phase trials have showed that several drugs presented an ideal curative effect, monotherapy cannot be entirely satisfactory in the treatment of cachexia-associated symptoms due to its complex and multifactorial pathogenesis. Therefore, the lack of definitive therapeutic strategies for cancer cachexia emphasizes the need to develop a better understanding of the underlying mechanisms. Increasing evidences show that the progression of cachexia is associated with metabolic alternations, which mainly include excessive energy expenditure, increased proteolysis and mitochondrial dysfunction. In this review, we provided an overview of the key mechanisms of cancer cachexia, with a major focus on muscle atrophy, adipose tissue wasting, anorexia and fatigue and updated the latest progress of pharmacological management of cancer cachexia, thereby further advancing the interventions that can counteract cancer cachexia.
Collapse
|
25
|
Lithium Chloride Protects against Sepsis-Induced Skeletal Muscle Atrophy and Cancer Cachexia. Cells 2021; 10:cells10051017. [PMID: 33925786 PMCID: PMC8146089 DOI: 10.3390/cells10051017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/19/2022] Open
Abstract
Inflammation-mediated skeletal muscle wasting occurs in patients with sepsis and cancer cachexia. Both conditions severely affect patient morbidity and mortality. Lithium chloride has previously been shown to enhance myogenesis and prevent certain forms of muscular dystrophy. However, to our knowledge, the effect of lithium chloride treatment on sepsis-induced muscle atrophy and cancer cachexia has not yet been investigated. In this study, we aimed to examine the effects of lithium chloride using in vitro and in vivo models of cancer cachexia and sepsis. Lithium chloride prevented wasting in myotubes cultured with cancer cell-conditioned media, maintained the expression of the muscle fiber contractile protein, myosin heavy chain 2, and inhibited the upregulation of the E3 ubiquitin ligase, Atrogin-1. In addition, it inhibited the upregulation of inflammation-associated cytokines in macrophages treated with lipopolysaccharide. In the animal model of sepsis, lithium chloride treatment improved body weight, increased muscle mass, preserved the survival of larger fibers, and decreased the expression of muscle-wasting effector genes. In a model of cancer cachexia, lithium chloride increased muscle mass, enhanced muscle strength, and increased fiber cross-sectional area, with no significant effect on tumor mass. These results indicate that lithium chloride exerts therapeutic effects on inflammation-mediated skeletal muscle wasting, such as sepsis-induced muscle atrophy and cancer cachexia.
Collapse
|
26
|
Kumagai H, Coelho AR, Wan J, Mehta HH, Yen K, Huang A, Zempo H, Fuku N, Maeda S, Oliveira PJ, Cohen P, Kim SJ. MOTS-c reduces myostatin and muscle atrophy signaling. Am J Physiol Endocrinol Metab 2021; 320:E680-E690. [PMID: 33554779 PMCID: PMC8238132 DOI: 10.1152/ajpendo.00275.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Obesity and type 2 diabetes are metabolic diseases, often associated with sarcopenia and muscle dysfunction. MOTS-c, a mitochondrial-derived peptide, acts as a systemic hormone and has been implicated in metabolic homeostasis. Although MOTS-c improves insulin sensitivity in skeletal muscle, whether MOTS-c impacts muscle atrophy is not known. Myostatin is a negative regulator of skeletal muscle mass and also one of the possible mediators of insulin resistance-induced skeletal muscle wasting. Interestingly, we found that plasma MOTS-c levels are inversely correlated with myostatin levels in human subjects. We further demonstrated that MOTS-c prevents palmitic acid-induced atrophy in differentiated C2C12 myotubes, whereas MOTS-c administration decreased myostatin levels in plasma in diet-induced obese mice. By elevating AKT phosphorylation, MOTS-c inhibits the activity of an upstream transcription factor for myostatin and other muscle wasting genes, FOXO1. MOTS-c increases mTORC2 and inhibits PTEN activity, which modulates AKT phosphorylation. Further upstream, MOTS-c increases CK2 activity, which leads to PTEN inhibition. These results suggest that through inhibition of myostatin, MOTS-c could be a potential therapy for insulin resistance-induced skeletal muscle atrophy as well as other muscle wasting phenotypes including sarcopenia.NEW & NOTEWORTHY MOTS-c, a mitochondrial-derived peptide reduces high-fat-diet-induced muscle atrophy signaling by reducing myostatin expression. The CK2-PTEN-mTORC2-AKT-FOXO1 pathways play key roles in MOTS-c action on myostatin expression.
Collapse
Affiliation(s)
- Hiroshi Kumagai
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Ana Raquel Coelho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal
| | - Junxiang Wan
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Hemal H Mehta
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Kelvin Yen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Amy Huang
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Hirofumi Zempo
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
- Department of Administrative Nutrition, Faculty of Health and Nutrition, Tokyo Seiei College, Tokyo, Japan
| | - Noriyuki Fuku
- Graduate School of Health and Sports Science, Juntendo University, Chiba, Japan
| | - Seiji Maeda
- Faculty of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Paulo J Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Cantanhede, Portugal
| | - Pinchas Cohen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| | - Su-Jeong Kim
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, California
| |
Collapse
|
27
|
Investigation into the role of anti-diabetic agents in cachexia associated with metastatic cancer. Life Sci 2021; 274:119329. [PMID: 33711389 DOI: 10.1016/j.lfs.2021.119329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/19/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
Cancer cachexia (CC) is a syndrome associated with cancer, and the global burden is increasing rapidly. Alteration in carbohydrate, lipid and protein metabolism along with systemic inflammation are characteristics of CC. Until now the available treatment for CC is limited to controlling inflammation and nutrition. Anti-diabetics are widely used agents to treat diabetics, this agent's act by regulating the carbohydrate metabolism, also they are known to have beneficial effects in maintaining protein and lipid balance. Role of anti-diabetics in cancer is being evaluated continuously and biguanides, dipeptidyl peptidase 4 (DPP4) inhibitors and Sodium glucose co-transporter 2 (SGLT2) inhibitors have proven anti-cancer potential. In this study, metastatic B16-F1 cell line induced cancer cachexia model used to evaluate potential of biguanides (metformin), DPP-4 inhibitors (teneligliptin and vildagliptin) and SGLT2 inhibitors (empagliflozin and dapagliflozin) in cancer cachexia. Our results suggest that anti-diabetic agents have potential to decrease rate of proliferation of tumor, restrict body mass markers, decrease inflammation, regulate carbohydrate mechanism and induce skeletal muscle hypertrophy. These findings may be helpful in management of cancer cachexia and increase the quality of life and survival chances of cancer cachexia patient.
Collapse
|
28
|
Berardi E, Madaro L, Lozanoska-Ochser B, Adamo S, Thorrez L, Bouche M, Coletti D. A Pound of Flesh: What Cachexia Is and What It Is Not. Diagnostics (Basel) 2021; 11:diagnostics11010116. [PMID: 33445790 PMCID: PMC7828214 DOI: 10.3390/diagnostics11010116] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/18/2022] Open
Abstract
Body weight loss, mostly due to the wasting of skeletal muscle and adipose tissue, is the hallmark of the so-called cachexia syndrome. Cachexia is associated with several acute and chronic disease states such as cancer, chronic obstructive pulmonary disease (COPD), heart and kidney failure, and acquired and autoimmune diseases and also pharmacological treatments such as chemotherapy. The clinical relevance of cachexia and its impact on patients’ quality of life has been neglected for decades. Only recently did the international community agree upon a definition of the term cachexia, and we are still awaiting the standardization of markers and tests for the diagnosis and staging of cancer-related cachexia. In this review, we discuss cachexia, considering the evolving use of the term for diagnostic purposes and the implications it has for clinical biomarkers, to provide a comprehensive overview of its biology and clinical management. Advances and tools developed so far for the in vitro testing of cachexia and drug screening will be described. We will also evaluate the nomenclature of different forms of muscle wasting and degeneration and discuss features that distinguish cachexia from other forms of muscle wasting in the context of different conditions.
Collapse
Affiliation(s)
- Emanuele Berardi
- Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium; (E.B.); (L.T.)
- Faculty of Rehabilitation Sciences, REVAL, Hasselt University (UHasselt), 3590 Diepenbeek, Belgium
| | - Luca Madaro
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (B.L.-O.); (S.A.); (D.C.)
| | - Biliana Lozanoska-Ochser
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (B.L.-O.); (S.A.); (D.C.)
| | - Sergio Adamo
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (B.L.-O.); (S.A.); (D.C.)
| | - Lieven Thorrez
- Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium; (E.B.); (L.T.)
| | - Marina Bouche
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (B.L.-O.); (S.A.); (D.C.)
- Correspondence: ; Tel.: +39-(6)-4976-6755/6573
| | - Dario Coletti
- DAHFMO Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy; (L.M.); (B.L.-O.); (S.A.); (D.C.)
- Biological Adaptation and Ageing, CNRS UMR 8256, Inserm U1164, Institut de Biologie Paris-Seine, Sorbonne Université, 75006 Paris, France
| |
Collapse
|
29
|
Lenehan PJ, Cirella A, Uchida AM, Crowley SJ, Sharova T, Boland G, Dougan M, Dougan SK, Heckler M. Type 2 immunity is maintained during cancer-associated adipose tissue wasting. IMMUNOTHERAPY ADVANCES 2021; 1:ltab011. [PMID: 34291232 PMCID: PMC8286632 DOI: 10.1093/immadv/ltab011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/21/2021] [Accepted: 05/30/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Cachexia is a systemic metabolic disorder characterized by loss of fat and muscle mass, which disproportionately impacts patients with gastrointestinal malignancies such as pancreatic cancer. While the immunologic shifts contributing to the development of other adipose tissue (AT) pathologies such as obesity have been well described, the immune microenvironment has not been studied in the context of cachexia. METHODS We performed bulk RNA-sequencing, cytokine arrays, and flow cytometry to characterize the immune landscape of visceral AT (VAT) in the setting of pancreatic and colorectal cancers. RESULTS The cachexia inducing factor IL-6 is strongly elevated in the wasting VAT of cancer bearing mice, but the regulatory type 2 immune landscape which characterizes healthy VAT is maintained. Pathologic skewing toward Th1 and Th17 inflammation is absent. Similarly, the VAT of patients with colorectal cancer is characterized by a Th2 signature with abundant IL-33 and eotaxin-2, albeit also with high levels of IL-6. CONCLUSIONS Wasting AT during the development of cachexia may not undergo drastic changes in immune composition like those seen in obese AT. Our approach provides a framework for future immunologic analyses of cancer associated cachexia.
Collapse
Affiliation(s)
- Patrick J Lenehan
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Assunta Cirella
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Amiko M Uchida
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Stephanie J Crowley
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Tatyana Sharova
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Genevieve Boland
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Max Heckler
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Surgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
30
|
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: 4.6] [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
|
31
|
Thaiss WM, Gatidis S, Sartorius T, Machann J, Peter A, Eigentler TK, Nikolaou K, Pichler BJ, Kneilling M. Noninvasive, longitudinal imaging-based analysis of body adipose tissue and water composition in a melanoma mouse model and in immune checkpoint inhibitor-treated metastatic melanoma patients. Cancer Immunol Immunother 2020; 70:1263-1275. [PMID: 33130917 PMCID: PMC8053172 DOI: 10.1007/s00262-020-02765-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022]
Abstract
Background As cancer cachexia (CC) is associated with cancer progression, early identification would be beneficial. The aim of this study was to establish a workflow for automated MRI-based segmentation of visceral (VAT) and subcutaneous adipose tissue (SCAT) and lean tissue water (LTW) in a B16 melanoma animal model, monitor diseases progression and transfer the protocol to human melanoma patients for therapy assessment. Methods For in vivo monitoring of CC B16 melanoma-bearing and healthy mice underwent longitudinal three-point DIXON MRI (days 3, 12, 17 after subcutaneous tumor inoculation). In a prospective clinical study, 18 metastatic melanoma patients underwent MRI before, 2 and 12 weeks after onset of checkpoint inhibitor therapy (CIT; n = 16). We employed an in-house MATLAB script for automated whole-body segmentation for detection of VAT, SCAT and LTW. Results B16 mice exhibited a CC phenotype and developed a reduced VAT volume compared to baseline (B16 − 249.8 µl, − 25%; controls + 85.3 µl, + 10%, p = 0.003) and to healthy controls. LTW was increased in controls compared to melanoma mice. Five melanoma patients responded to CIT, 7 progressed, and 6 displayed a mixed response. Responding patients exhibited a very limited variability in VAT and SCAT in contrast to others. Interestingly, the LTW was decreased in CIT responding patients (− 3.02% ± 2.67%; p = 0.0034) but increased in patients with progressive disease (+ 1.97% ± 2.19%) and mixed response (+ 4.59% ± 3.71%). Conclusion MRI-based segmentation of fat and water contents adds essential additional information for monitoring the development of CC in mice and metastatic melanoma patients during CIT or other treatment approaches.
Collapse
Affiliation(s)
- Wolfgang M Thaiss
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany.,Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany.,Department of Nuclear Medicine, University of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany.,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany
| | - Tina Sartorius
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany.,Section of Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peter
- German Center for Diabetes Research (DZD E.V.), Neuherberg, Germany.,Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany.,Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, Tübingen, Germany
| | - Thomas K Eigentler
- Department of Dermatology, University Hospital Tübingen, Liebermeisterstreet 20, 72076, Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, Eberhard Karls University, 72076, Tübingen, Germany
| | - Bernd J Pichler
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany.,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) Partner Site Tübingen, 72076, Tübingen, Germany
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, 72076, Tübingen, Germany. .,iFIT-Cluster of Excellence, Eberhard Karls University, 72076, Tübingen, Germany. .,Department of Dermatology, University Hospital Tübingen, Liebermeisterstreet 20, 72076, Tübingen, Germany.
| |
Collapse
|
32
|
Santos IM, Mendes L, Carolino E, Santos CA. Nutritional Status, Functional Status, and Quality of Life - What is the Impact and Relationship on Cancer Patients? Nutr Cancer 2020; 73:2554-2567. [PMID: 33121266 DOI: 10.1080/01635581.2020.1839520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
RATIONALE AND AIMS Malnutrition is common and multifactorial in cancer patients (CP), combining the systemic inflammatory process with decreased food intake, loss of muscle and bone mass, and decreased functional status (FS). We aimed to track and evaluate the nutritional status (NS) of CP; associate results between hospitalized patients (HP) and day hospital patients (DHP); associate NS with tumor, symptoms, and FS. METHODS Cross-sectional observational study in HP or DHP from Garcia de Orta Hospital, over 18 years old. NS was monitored and evaluated using Nutritional Risk Screening (NRS-2002), Patient-Generated Subjective Global Assessment (PG-SGA), and anthropometric and biochemical parameters. To assess FS we used the Eastern Cooperative Oncology Group (ECOG), Karnofsky Performance Scale Index (KPSI), and handgrip dynamometer (HGD). RESULTS The 265 CP (114-HP, 151-DHP), of which 34.2%-HP and 17.2%-DHP had low BMI. From NRS-2002, 86.0% and 35.8% were respectively at nutritional risk. Using PG-SGA, 93.0% and 39.7% were respectively malnourished. PG-SGA were positively correlated with ECOG (p < 0.01) and negatively correlated with KPSI (p < 0.01), BMI (p < 0.01), and handgrip strength-HGS (p < 0.01-DHP and p < 0.05-HP). CONCLUSIONS PG-SGA and FS scales are appropriate and validated tools for early identification of malnutrition and FS in CP. HGD can be a useful tool for assessing FS and NS.
Collapse
Affiliation(s)
- Inês Miranda Santos
- Dietetics and Nutrition, Lisbon School of Health Technology, Lisbon, Portugal
| | - Lino Mendes
- Dietetics and Nutrition, Lisbon School of Health Technology, Lisbon, Portugal.,H&TRC (Health and Technology Research Center), Lisbon, Portugal
| | - Elisabete Carolino
- Department of Natural and Exact Sciences - Mathematical Science Area, Lisbon School of Health Technology, Lisbon, Portugal
| | - Carla Adriana Santos
- H&TRC (Health and Technology Research Center), Lisbon, Portugal.,Nutrition Department & GENE (Enteral Feeding Team), Garcia de Orta Hospital, Lisbon, Portugal
| |
Collapse
|
33
|
Turbitt WJ, Orlandella RM, Gibson JT, Peterson CM, Norian LA. Therapeutic Time-restricted Feeding Reduces Renal Tumor Bioluminescence in Mice but Fails to Improve Anti-CTLA-4 Efficacy. Anticancer Res 2020; 40:5445-5456. [PMID: 32988866 PMCID: PMC7957951 DOI: 10.21873/anticanres.14555] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/24/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND/AIM Dietary interventions like time-restricted feeding (TRF) show promising anti-cancer properties. We examined whether therapeutic TRF alone or combined with immunotherapy would diminish renal tumor growth in mice of varying body weights. MATERIALS AND METHODS Young (7 week) chow-fed or older (27 week) high-fat diet (HFD)-fed BALB/c mice were orthotopically injected with renal tumor cells expressing luciferase. After tumor establishment, mice were randomized to ad libitum feeding or TRF +/- anti-CTLA-4. Body composition, tumor viability and growth, and immune responses were quantified. RESULTS TRF alone reduced renal tumor bioluminescence in older HFD-fed, but not young chow-fed mice. In the latter, TRF mitigated tumor-induced loss of lean- and fat-mass. However, TRF did not alter excised renal tumor weights or intratumoral immune responses and failed to improve anti-CTLA-4 outcomes in any mice. CONCLUSION Therapeutic TRF exhibits modest anti-cancer properties but fails to improve anti-CTLA-4 immune checkpoint blockade in murine renal cancer.
Collapse
Affiliation(s)
- William J Turbitt
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Rachael M Orlandella
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Justin T Gibson
- Graduate Biomedical Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Courtney M Peterson
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| | - Lyse A Norian
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, U.S.A.
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, U.S.A
| |
Collapse
|
34
|
McKeaveney C, Maxwell P, Noble H, Reid J. A Critical Review of Multimodal Interventions for Cachexia. Adv Nutr 2020; 12:523-532. [PMID: 32970097 PMCID: PMC8262513 DOI: 10.1093/advances/nmaa111] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/18/2020] [Accepted: 08/14/2020] [Indexed: 12/17/2022] Open
Abstract
Currently, there are no standardized treatments for cachexia or severe wasting. There is a growing consensus advocating multimodal interventions to address the complex pathogenesis and metabolic alterations in these conditions. This review examined multimodal treatments intended to alleviate and/or stabilize cachexia and severe wasting. The objectives of this review were to 1) identify multimodal interventions for the treatment of cachexia or associated wasting syndromes in patients with a chronic illness, 2) assess the quality of these studies, and 3) assess the effectiveness of multimodal interventions. Electronic databases including PubMed, MEDLINE, EMBASE, Scopus, Web of Science, Cochrane Library, CINAHL, PEDro, OpenGrey, and clinicaltrials.org were systematically searched using both text words and MeSH (medical subject heading) terms. The literature revealed a dearth of large, well-conducted trials in this area. Fourteen trials (n = 5 cancer, n = 5 chronic obstructive pulmonary disease, n = 4 chronic kidney disease) were included in this review. A total of 1026 patients were included across all studies; sample size ranged between 21 and 138 patients. Baseline and follow-up data were collected between 6 wk and 24 mo. All demonstrated some improvement in favor of the treatment groups, in relevant measures of body composition, nutrition, biomarkers, and functionality; however, caution should be applied due to the heterogenous nature of the interventions and small sample sizes. Overall, the evidence from this review supports the role of multimodal interventions in the treatment of severe wasting. However, randomized controlled trials with a powered sample size and sufficiently lengthy interaction period are necessary to assess if multimodal interventions are effective forms of therapy for improving body composition and nutritional and physical status in patients with cachexia and wasting. The protocol for this review is registered with Prospero (ID: CRD42019124374).
Collapse
Affiliation(s)
- Clare McKeaveney
- School of Nursing and Midwifery, Medical Biology Centre, Queen's University
Belfast, Belfast, Northern Ireland
| | - Peter Maxwell
- Centre for Public Health, Queen's University Belfast, Institute of Clinical
Science, Royal Victoria Hospital, Belfast, Northern Ireland,Regional Nephrology Unit, Belfast City Hospital, Belfast Health Social Care
Trust, Belfast, Northern Ireland
| | - Helen Noble
- School of Nursing and Midwifery, Medical Biology Centre, Queen's University
Belfast, Belfast, Northern Ireland
| | | |
Collapse
|
35
|
Shakri AR, James Zhong T, Ma W, Coker C, Hegde R, Scholze H, Chin V, Szabolcs M, Hibshoosh H, Tanji K, Baer R, Kumar Biswas A, Acharyya S. Aberrant Zip14 expression in muscle is associated with cachexia in a Bard1-deficient mouse model of breast cancer metastasis. Cancer Med 2020; 9:6766-6775. [PMID: 32730698 PMCID: PMC7520359 DOI: 10.1002/cam4.3242] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/30/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022] Open
Abstract
Nearly 80% of advanced cancer patients are afflicted with cachexia, a debilitating syndrome characterized by extensive loss of muscle mass and function. Cachectic cancer patients have a reduced tolerance to antineoplastic therapies and often succumb to premature death from the wasting of respiratory and cardiac muscles. Since there are no available treatments for cachexia, it is imperative to understand the mechanisms that drive cachexia in order to devise effective strategies to treat it. Although 25% of metastatic breast cancer patients develop symptoms of muscle wasting, mechanistic studies of breast cancer cachexia have been hampered by a lack of experimental models. Using tumor cells deficient for BARD1, a subunit of the BRCA1/BARD1 tumor suppressor complex, we have developed a new orthotopic model of triple‐negative breast cancer that spontaneously metastasizes to the lung and leads to systemic muscle deterioration. We show that expression of the metal‐ion transporter, Zip14, is markedly upregulated in cachectic muscles from these mice and is associated with elevated intramuscular zinc and iron levels. Aberrant Zip14 expression and altered metal‐ion homeostasis could therefore represent an underlying mechanism of cachexia development in human patients with triple‐negative breast cancer. Our study provides a unique model for studying breast cancer cachexia and identifies a potential therapeutic target for its treatment.
Collapse
Affiliation(s)
| | - Timothy James Zhong
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.,Graduate School of Arts and Sciences, Department of Pathobiology and Mechanisms of Disease, Columbia University Irving Medical Center, New York, NY, USA
| | - Wanchao Ma
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Courtney Coker
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Rohaan Hegde
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Hanna Scholze
- Barnard College, Columbia University, New York, NY, USA.,Weill Cornell Medical College, New York, NY, USA
| | - Vanessa Chin
- Barnard College, Columbia University, New York, NY, USA
| | - Matthias Szabolcs
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Kurenai Tanji
- Division of Neuropathology, Department of Pathology and Cell Biology, Columbia University Medical Center and New York Presbyterian Hospital, New York, NY, USA
| | - Richard Baer
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Anup Kumar Biswas
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Swarnali Acharyya
- Institute for Cancer Genetics, Columbia University, New York, NY, USA.,Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.,Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| |
Collapse
|
36
|
Peixoto da Silva S, Santos JMO, Costa E Silva MP, Gil da Costa RM, Medeiros R. Cancer cachexia and its pathophysiology: links with sarcopenia, anorexia and asthenia. J Cachexia Sarcopenia Muscle 2020; 11:619-635. [PMID: 32142217 PMCID: PMC7296264 DOI: 10.1002/jcsm.12528] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/07/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by a progressive loss of skeletal muscle mass, along with adipose tissue wasting, systemic inflammation and other metabolic abnormalities leading to functional impairment. Cancer cachexia has long been recognized as a direct cause of complications in cancer patients, reducing quality of life and worsening disease outcomes. Some related conditions, like sarcopenia (age-related muscle wasting), anorexia (appetite loss) and asthenia (reduced muscular strength and fatigue), share some key features with cancer cachexia, such as weakness and systemic inflammation. Understanding the interplay and the differences between these conditions is critical to advance basic and translational research in this field, improving the accuracy of diagnosis and contributing to finally achieve effective therapies for affected patients.
Collapse
Affiliation(s)
- Sara Peixoto da Silva
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Joana M O Santos
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Maria Paula Costa E Silva
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.,Palliative Care Service, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Rui M Gil da Costa
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Center for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal.,Postgraduate Programme in Adult Health (PPGSAD) and Tumour Biobank, Federal University of Maranhão (UFMA), São Luís, Brazil
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.,Virology Service, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal.,Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, Porto, Portugal.,Research Department, Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), Porto, Portugal
| |
Collapse
|
37
|
Abstract
During nearly 100 years of research on cancer cachexia (CC), science has been reciting the same mantra: it is a multifactorial syndrome. The aim of this paper is to show that the symptoms are many, but they have a single cause: anoxia. CC is a complex and devastating condition that affects a high proportion of advanced cancer patients. Unfortunately, it cannot be reversed by traditional nutritional support and it generally reduces survival time. It is characterized by significant weight loss, mainly from fat deposits and skeletal muscles. The occurrence of cachexia in cancer patients is usually a late phenomenon. The conundrum is why do similar patients with similar tumors, develop cachexia and others do not? Even if cachexia is mainly a metabolic dysfunction, there are other issues involved such as the activation of inflammatory responses and crosstalk between different cell types. The exact mechanism leading to a wasting syndrome is not known, however there are some factors that are surely involved, such as anorexia with lower calorie intake, increased glycolytic flux, gluconeogenesis, increased lipolysis and severe tumor hypoxia. Based on this incomplete knowledge we put together a scheme explaining the molecular mechanisms behind cancer cachexia, and surprisingly, there is one cause that explains all of its characteristics: anoxia. With this different view of CC we propose a treatment based on the physiopathology that leads from anoxia to the symptoms of CC. The fundamentals of this hypothesis are based on the idea that CC is the result of anoxia causing intracellular lactic acidosis. This is a dangerous situation for cell survival which can be solved by activating energy consuming gluconeogenesis. The process is conducted by the hypoxia inducible factor-1α. This hypothesis was built by putting together pieces of evidence produced by authors working on related topics.
Collapse
|
38
|
Biswas AK, Acharyya S. Cancer-Associated Cachexia: A Systemic Consequence of Cancer Progression. ANNUAL REVIEW OF CANCER BIOLOGY 2020; 4:391-411. [DOI: 10.1146/annurev-cancerbio-030419-033642] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
Cancer is a life-threatening disease that has plagued humans for centuries. The vast majority of cancer-related mortality results from metastasis. Indeed, the invasive growth of metastatic cancer cells in vital organs causes fatal organ dysfunction, but metastasis-related deaths also result from cachexia, a debilitating wasting syndrome characterized by an involuntary loss of skeletal muscle mass and function. In fact, about 80% of metastatic cancer patients suffer from cachexia, which often renders them too weak to tolerate standard doses of anticancer therapies and makes them susceptible to death from cardiac and respiratory failure. The goals of this review are to highlight important findings that help explain how cancer-induced systemic changes drive the development of cachexia and to discuss unmet challenges and potential therapeutic strategies targeting cachexia to improve the quality of life and survival of cancer patients.
Collapse
Affiliation(s)
- Anup K. Biswas
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Swarnali Acharyya
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Pathology and Cell Biology and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
39
|
Upregulation of ZIP14 and Altered Zinc Homeostasis in Muscles in Pancreatic Cancer Cachexia. Cancers (Basel) 2019; 12:cancers12010003. [PMID: 31861290 PMCID: PMC7016633 DOI: 10.3390/cancers12010003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/04/2019] [Accepted: 12/13/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal cancer type in which the mortality rate approaches the incidence rate. More than 85% of PDAC patients experience a profound loss of muscle mass and function, known as cachexia. PDAC patients with this condition suffer from decreased tolerance to anti-cancer therapies and often succumb to premature death due to respiratory and cardiac muscle wasting. Yet, there are no approved therapies available to alleviate cachexia. We previously found that upregulation of the metal ion transporter, Zip14, and altered zinc homeostasis are critical mediators of cachexia in metastatic colon, lung, and breast cancer models. Here, we show that a similar mechanism is likely driving the development of cachexia in PDAC. In two independent experimental metastasis models generated from the murine PDAC cell lines, Pan02 and FC1242, we observed aberrant Zip14 expression and increased zinc ion levels in cachectic muscles. Moreover, in advanced PDAC patients, high levels of ZIP14 in muscles correlated with the presence of cachexia. These studies underscore the importance of altered ZIP14 function in PDAC-associated cachexia development and highlight a potential therapeutic opportunity for improving the quality of life and prolonging survival in PDAC patients.
Collapse
|
40
|
Lee H, Heo JW, Kim AR, Kweon M, Nam S, Lim JS, Sung MK, Kim SE, Ryu JH. Z-ajoene from Crushed Garlic Alleviates Cancer-Induced Skeletal Muscle Atrophy. Nutrients 2019; 11:nu11112724. [PMID: 31717643 PMCID: PMC6893518 DOI: 10.3390/nu11112724] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 11/05/2019] [Accepted: 11/07/2019] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle atrophy is one of the major symptoms of cancer cachexia. Garlic (Allium sativum), one of the world's most commonly used and versatile herbs, has been employed for the prevention and treatment of diverse diseases for centuries. In the present study, we found that ajoene, a sulfur compound found in crushed garlic, exhibits protective effects against muscle atrophy. Using CT26 tumor-bearing BALB/c mice, we demonstrate in vivo that ajoene extract alleviated muscle degradation by decreasing not only myokines secretion but also janus kinase/signal transducer and activator of transcription 3 (JAK/STAT3) and SMADs/forkhead box (FoxO) signaling pathways, thereby suppressing muscle-specific E3 ligases. In mouse skeletal myoblasts, Z-ajoene enhanced myogenesis as evidenced by increased expression of myogenic markers via p38 mitogen-activated protein kinase (MAPK) activation. In mature myotubes, Z-ajoene protected against muscle protein degradation induced by conditioned media from CT26 colon carcinoma cells, by suppressing expression of muscle specific E3 ligases and nuclear transcription factor kappa B (NF-κB) phosphorylation which contribute to muscle atrophy. Moreover, Z-ajoene treatment improved myofiber formation via stimulation of muscle protein synthesis. These findings suggest that ajoene extract and Z-ajoene can attenuate skeletal muscle atrophy induced by cancer cachexia through suppressing inflammatory responses and the muscle wasting as well as by promoting muscle protein synthesis.
Collapse
Affiliation(s)
- Hyejin Lee
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
| | - Ji-Won Heo
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - A-Reum Kim
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - Minson Kweon
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
| | - Sorim Nam
- Division of Biological Sciences and Cellular Heterogeneity Research Center, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (S.N.); (J.-S.L.)
| | - Jong-Seok Lim
- Division of Biological Sciences and Cellular Heterogeneity Research Center, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (S.N.); (J.-S.L.)
| | - Mi-Kyung Sung
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
| | - Sung-Eun Kim
- Department of Food and Nutrition, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (J.-W.H.); (A.-R.K.); (M.-K.S.)
- Correspondence: (S.-E.K.); (J.-H.R.); Tel.: +82-2-2077-7722 (S.-E.K.); +82-2-710-9568 (J.-H.R.)
| | - Jae-Ha Ryu
- Research Institute of Pharmaceutical Sciences, Sookmyung Women’s University, Yongsan-Gu, Seoul 04310, Korea; (H.L.); minson-_-@nate.com (M.K.)
- Correspondence: (S.-E.K.); (J.-H.R.); Tel.: +82-2-2077-7722 (S.-E.K.); +82-2-710-9568 (J.-H.R.)
| |
Collapse
|
41
|
Cachexia Anorexia Syndrome and Associated Metabolic Dysfunction in Peritoneal Metastasis. Int J Mol Sci 2019; 20:ijms20215444. [PMID: 31683709 PMCID: PMC6862625 DOI: 10.3390/ijms20215444] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022] Open
Abstract
Patients with peritoneal metastasis (PM) of gastrointestinal and gynecological origin present with a nutritional deficit characterized by increased resting energy expenditure (REE), loss of muscle mass, and protein catabolism. Progression of peritoneal metastasis, as with other advanced malignancies, is associated with cancer cachexia anorexia syndrome (CAS), involving poor appetite (anorexia), involuntary weight loss, and chronic inflammation. Eventual causes of mortality include dysfunctional metabolism and energy store exhaustion. Etiology of CAS in PM patients is multifactorial including tumor growth, host response, cytokine release, systemic inflammation, proteolysis, lipolysis, malignant small bowel obstruction, ascites, and gastrointestinal side effects of drug therapy (chemotherapy, opioids). Metabolic changes of CAS in PM relate more to a systemic inflammatory response than an adaptation to starvation. Metabolic reprogramming is required for cancer cells shed into the peritoneal cavity to resist anoikis (i.e., programmed cell death). Profound changes in hexokinase metabolism are needed to compensate ineffective oxidative phosphorylation in mitochondria. During the development of PM, hypoxia inducible factor-1α (HIF-1α) plays a key role in activating both aerobic and anaerobic glycolysis, increasing the uptake of glucose, lipid, and glutamine into cancer cells. HIF-1α upregulates hexokinase II, phosphoglycerate kinase 1 (PGK1), pyruvate dehydrogenase kinase (PDK), pyruvate kinase muscle isoenzyme 2 (PKM2), lactate dehydrogenase (LDH) and glucose transporters (GLUT) and promotes cytoplasmic glycolysis. HIF-1α also stimulates the utilization of glutamine and fatty acids as alternative energy substrates. Cancer cells in the peritoneal cavity interact with cancer-associated fibroblasts and adipocytes to meet metabolic demands and incorporate autophagy products for growth. Therapy of CAS in PM is challenging. Optimal nutritional intake alone including total parenteral nutrition is unable to reverse CAS. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) stabilized nutritional status in a significant proportion of PM patients. Agents targeting the mechanisms of CAS are under development.
Collapse
|
42
|
Dijk FJ, van Dijk M, Dorresteijn B, van Norren K. DPA shows comparable chemotherapy sensitizing effects as EPA upon cellular incorporation in tumor cells. Oncotarget 2019; 10:5983-5992. [PMID: 31666929 PMCID: PMC6800265 DOI: 10.18632/oncotarget.27236] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Dietary supplementation with ω-3 polyunsaturated fatty acids (PUFAs) has been reported to enhance the sensitivity of tumor cells towards chemotherapy. Most enhancing effects are described for ω-3 PUFAs EPA and DHA; less evidence is available with the intermediate DPA. We studied the chemotherapy enhancing effects of EPA, DPA and DHA in murine colon C26 adenocarcinoma cells and showed that DPA displayed similar chemosensitizing effects as EPA. Moreover, EPA supplementation increased cellular DPA content. In a C26 tumor-bearing mouse model, we studied the incorporation of ω-3 PUFA in tumor and skeletal muscle after a diet with different ω-3 PUFA sources. Although little DPA was present in the fatty acid food sources, in those that contained considerable EPA concentrations, DPA levels were higher in tumor and muscle tissue. From these studies, we conclude that EPA and DPA show chemosensitizing effects and that intake of EPA or EPA-containing nutrition leads to increased cellular DPA content by elongation. These findings support the use of ω-3 PUFA containing nutritional supplementations in cancer patients during chemotherapy treatment.
Collapse
Affiliation(s)
- Francina J Dijk
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Miriam van Dijk
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Bram Dorresteijn
- Danone Nutricia Research, Nutricia Advanced Medical Nutrition, Utrecht, The Netherlands
| | - Klaske van Norren
- Nutritional Biology, Department of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
43
|
Growth Hormone Secretagogues and the Regulation of Calcium Signaling in Muscle. Int J Mol Sci 2019; 20:ijms20184361. [PMID: 31491959 PMCID: PMC6769538 DOI: 10.3390/ijms20184361] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/02/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022] Open
Abstract
Growth hormone secretagogues (GHS) are a family of synthetic molecules, first discovered in the late 1970s for their ability to stimulate growth hormone (GH) release. Many effects of GHS are mediated by binding to GHS-R1a, the receptor for the endogenous hormone ghrelin, a 28-amino acid peptide isolated from the stomach. Besides endocrine functions, both ghrelin and GHS are endowed with some relevant extraendocrine properties, including stimulation of food intake, anticonvulsant and anti-inflammatory effects, and protection of muscle tissue in different pathological conditions. In particular, ghrelin and GHS inhibit cardiomyocyte and endothelial cell apoptosis and improve cardiac left ventricular function during ischemia–reperfusion injury. Moreover, in a model of cisplatin-induced cachexia, GHS protect skeletal muscle from mitochondrial damage and improve lean mass recovery. Most of these effects are mediated by GHS ability to preserve intracellular Ca2+ homeostasis. In this review, we address the muscle-specific protective effects of GHS mediated by Ca2+ regulation, but also highlight recent findings of their therapeutic potential in pathological conditions characterized by skeletal or cardiac muscle impairment.
Collapse
|