1
|
Zhan C, Zhu Y, Fok MR, Jin L, Han B, Lin Y. Proteome-Wide Mendelian Randomisation Identifies Causal Links of Plasma Proteins With Periodontitis. Int Dent J 2024; 74:1258-1265. [PMID: 38729796 DOI: 10.1016/j.identj.2024.04.019] [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: 03/01/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE Periodontitis is a complex and multifactorial disease and it is challenging to decipher its underlying causes and mechanisms. This study attempted to explore potential circulating proteins in connection to periodontitis through proteome-wide Mendelian randomisation (MR). METHODS We analysed 1722 circulating proteins to identify prospective drug targets for tackling periodontitis, using the genomic dataset from the FinnGen study. Two-sample MR was conducted to evaluate the bidirectional relationship between circulating proteins and periodontitis risk. A dataset from the UK Biobank was used to validate the findings. Single-cell analysis was performed to assess the cellular expression of the identified proteins within gingival tissues. RESULTS MR analyses found that genetically predicted circulating levels of von Willebrand factor A domain-containing 1 (von Willebrand factor A domain containing 1 [VWA1], odds ratios: 0.94, 95% CI 0.92-0.97, P = 1.28 × 10-5) were inversely associated with periodontitis. In contrast, the level of growth differentiation factor 15 (growth differentiation factor 15 [GDF15], odds ratios: 1.05, 95% CI 1.02-1.07, P = 2.12 × 10-5) might be associated with an increased risk of periodontitis. Single-cell analysis indicated that VWA1 was primarily expressed in endothelial cells of healthy gingival tissues, while the main source of GDF15 was not derived from periodontal cells. CONCLUSIONS The present study suggests that certain plasma proteins like VWA1 and GDF15 may be potentially indicative of the risk and susceptibility to periodontitis. These proteins could possibly be the potential therapeutic targets for treating periodontitis, and further investigation is highly warranted.
Collapse
Affiliation(s)
- Chaoning Zhan
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Yuexin Zhu
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Melissa Rachel Fok
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lijian Jin
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Bing Han
- Department of Orthodontics, Cranial-Facial Growth and Development Center, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Yifan Lin
- Division of Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Li L, Wazir J, Huang Z, Wang Y, Wang H. A comprehensive review of animal models for cancer cachexia: Implications for translational research. Genes Dis 2024; 11:101080. [PMID: 39220755 PMCID: PMC11364047 DOI: 10.1016/j.gendis.2023.101080] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 09/04/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by progressive weight loss and a disease process that nutritional support cannot reverse. Although progress has been made in preclinical research, there is still a long way to go in translating research findings into clinical practice. One of the main reasons for this is that existing preclinical models do not fully replicate the conditions seen in clinical patients. Therefore, it is important to understand the characteristics of existing preclinical models of cancer cachexia and pay close attention to the latest developments in preclinical models. The main models of cancer cachexia used in current research are allogeneic and xenograft models, genetically engineered mouse models, chemotherapy drug-induced models, Chinese medicine spleen deficiency models, zebrafish and Drosophila models, and cellular models. This review aims to revisit and summarize the commonly used animal models of cancer cachexia by evaluating existing preclinical models, to provide tools and support for translational medicine research.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
3
|
van de Lisdonk D, Li B. The area postrema: a critical mediator of brain-body interactions. Genes Dev 2024; 38:793-797. [PMID: 39362783 PMCID: PMC11535157 DOI: 10.1101/gad.352276.124] [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] [Indexed: 10/05/2024]
Abstract
The dorsal vagal complex contains three structures: the area postrema, the nucleus tractus solitarii, and the dorsal motor nucleus of the vagus. These structures are tightly linked, both anatomically and functionally, and have important yet distinct roles in not only conveying peripheral bodily signals to the rest of the brain but in the generation of behavioral and physiological responses. Reports on the new discoveries in these structures were highlights of the symposium. In this outlook, we focus on the roles of the area postrema in mediating brain-body interactions and its potential utility as a therapeutic target, especially in cancer cachexia.
Collapse
Affiliation(s)
- Daniëlle van de Lisdonk
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
- Center for Neuroscience, University of Amsterdam, Amsterdam 1098 XH, the Netherlands
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724, USA;
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou 310024, Zhejiang, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| |
Collapse
|
4
|
Kumar K, Bosch K, Vemuri V, Kratholm N, Rane M, Kakar SS. Withaferin A ameliorates ovarian cancer-induced renal damage through the regulation of expression of inflammatory cytokines. J Ovarian Res 2024; 17:199. [PMID: 39394174 PMCID: PMC11468018 DOI: 10.1186/s13048-024-01519-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Cachexia a multifactorial syndrome is a common sequala in patients with cancer. It varies from 42 to 80% depending upon the oncological stage and is directly responsible for 30% of deaths in these patients. Previous research from our laboratory demonstrated that peritoneal ovarian cancer generated in NSG mice resulted in skeletal and cardiac muscle atrophy - leading to loss of skeletal muscle mass and strength, and cardiac dysfunction (cachexia). Treatment of mice bearing i.p. tumors with withaferin A (WFA) showed reversal of skeletal muscle and cardiac cachexia. The present study is focused on determining effects of peritoneal ovarian tumors on kidney damage and effects of WFA treatment on ameliorating kidney damage. METHODS We generated intraperitoneal ovarian cancer by injecting female NSG mice with ovarian cancer cell line (A2780). After one week of injecting cancer cells, mice were treated with WFA (4 mg/kg) every third day, for three weeks. After 4 weeks of injection of cancer cells, the mice were sacrificed and various tissues including kidney and blood were collected, snap-frozen in liquid nitrogen, and stored at -800C. The presence of kidney biomarker creatinine, was measured in the plasma by an ELISA. The mRNA was isolated from mouse kidneys and was used to examine the expression levels of signaling proteins, inflammatory cytokines, and genes responsible for inducing cachexia (IL-1β, IL-6, TNF-α, TGF-β, GDF-15, and MYD88). RESULTS Our results showed a significant increase in levels of expression of inflammatory cytokine IL-1 β (p < 0.01), IL-6 (p < 0.001), TNF-α (p < 0.001), and other related genes including TRAF6 (p < 0.01), MYD88 (p < 0.01), and GDF-15 (p = 0.005) in tumor-bearing mice compared to controls. Treatment of mice bearing tumors with WFA attenuated the increase in expression of each gene. In addition, our results showed a significant increase in creatinine levels in circulation in tumor-bearing mice compared to control mice. Treatment of tumor-bearing mice with WFA resulted in a significant decrease in plasma creatinine levels compared to tumor-bearing mice. CONCLUSIONS Our results conclude that ovarian tumors in NSG mice caused kidney damage and renal dysfunction, which was effectively ameliorated by WFA treatment, suggesting a protective effect of WFA on kidney injury induced by ovarian cancer.
Collapse
Affiliation(s)
- Kusum Kumar
- Deparment of Biology, University of Louisville, Louisville, KY, USA
| | - Katherine Bosch
- Department of Biology and Chemistry, Liberty University, Lynchburg, VA, USA
| | - Vasa Vemuri
- Deparment of Biology, University of Louisville, Louisville, KY, USA
| | - Nicholas Kratholm
- Department of Physiology, School of Medicine, University of Louisville, 500 South Floyd Street, Louisville, KY, 40202, USA
| | - Madhavi Rane
- Department of Medicine, Division Nephrology, University of Louisville, Louisville, KY, USA
| | - Sham S Kakar
- Department of Physiology, School of Medicine, University of Louisville, 500 South Floyd Street, Louisville, KY, 40202, USA.
- Department of Medicine, Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
5
|
Li Y, Zhang J, Chen S, Ke Y, Li Y, Chen Y. Growth differentiation factor 15: Emerging role in liver diseases. Cytokine 2024; 182:156727. [PMID: 39111112 DOI: 10.1016/j.cyto.2024.156727] [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: 06/05/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/25/2024]
Abstract
Growth differentiation factor 15 (GDF15) is a cell stress-response cytokine within the transforming growth factor-β (TGFβ) superfamily. It is known to exert diverse effects on many metabolic pathways through its receptor GFRAL, which is expressed in the hindbrain, and transduces signals through the downstream receptor tyrosine kinase Ret. Since the liver is the core organ of metabolism, summarizing the functions of GDF15 is highly important. In this review, we assessed the relevant literature regarding the main metabolic, inflammatory, fibrogenic, tumorigenic and other effects of GDF15 on different liver diseases, including Metabolic dysfunction-associated steatotic liver disease(MASLD), alcohol and drug-induced liver injury, as well as autoimmune and viral hepatitis, with a particular focus on the pathogenesis of MASLD progression from hepatic steatosis to MASH, liver fibrosis and even hepatocellular carcinoma (HCC). Finally, we discuss the prospects of the clinical application potential of GDF15 along with its research and development progress. With better knowledge of GDF15, increasing in-depth research will lead to a new era in the field of liver diseases.
Collapse
Affiliation(s)
- Yu Li
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shurong Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yini Ke
- Department of Rheumatology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Youming Li
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yi Chen
- Department of Gastroenterology, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
6
|
Wang Q, Farhadipour M, Thijs T, Ruilova Sosoranga E, Van der Schueren B, Ceulemans LJ, Deleus E, Lannoo M, Tack J, Depoortere I. Bitter-tasting drugs tune GDF15 and GLP-1 expression via bitter taste or motilin receptors in the intestine of patients with obesity. Mol Metab 2024; 88:102002. [PMID: 39111389 PMCID: PMC11380393 DOI: 10.1016/j.molmet.2024.102002] [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: 06/26/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/22/2024] Open
Abstract
OBJECTIVE Growth differentiation factor 15 (GDF15), a stress related cytokine, was recently identified as a novel satiety signal acting via the GFRAL receptor located in the hindbrain. Bitter compounds are known to induce satiety via the release of glucagon-like peptide 1 (GLP-1) through activation of bitter taste receptors (TAS2Rs, 25 subtypes) on enteroendocrine cells in the gut. This study aimed to investigate whether and how bitter compounds induce a stress response in intestinal epithelial cells to affect GDF15 expression in patients with obesity, thereby facilitating satiety signaling from the gut. METHODS The acute effect of oral intake of the bitter-containing medication Plaquenil (hydroxychloroquine sulfate) on plasma GDF15 levels was evaluated in a placebo-controlled, double-blind, randomized, two-visit crossover study in healthy volunteers. Primary crypts isolated from the jejunal mucosa from patients with obesity were stimulated with vehicle or bitter compounds, and the effect on GDF15 expression was evaluated using RT-qPCR or ELISA. Immunofluorescence colocalization studies were performed between GDF15, epithelial cell type markers and TAS2Rs. The role of TAS2Rs was tested by 1) pretreatment with a TAS2R antagonist, GIV3727; 2) determining TAS2R4/43 polymorphisms that affect taste sensitivity to TAS2R4/43 agonists. RESULTS Acute intake of hydroxychloroquine sulfate increased GDF15 plasma levels, which correlated with reduced hunger scores and plasma ghrelin levels in healthy volunteers. This effect was mimicked in primary jejunal cultures from patients with obesity. GDF15 was expressed in enteroendocrine and goblet cells with higher expression levels in patients with obesity. Various bitter-tasting compounds (medicinal, plant extracts, bacterial) either increased or decreased GDF15 expression, with some also affecting GLP-1. The effect was mediated by specific intestinal TAS2R subtypes and the unfolded protein response pathway. The bitter-induced effect on GDF15/GLP-1 expression was influenced by the existence of TAS2R4 amino acid polymorphisms and TAS2R43 deletion polymorphisms that may predict patient's therapeutic responsiveness. However, the effect of the bitter-tasting antibiotic azithromycin on GDF15 release was mediated via the motilin receptor, possibly explaining some of its aversive side effects. CONCLUSIONS Bitter chemosensory and pharmacological receptors regulate the release of GDF15 from human gut epithelial cells and represent potential targets for modulating metabolic disorders or cachexia.
Collapse
Affiliation(s)
- Qian Wang
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Mona Farhadipour
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Theo Thijs
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | | - Bart Van der Schueren
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical and Experimental Endocrinology, University of Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Leuven Intestinal Failure and Transplantation (LIFT) Center, University Hospitals Leuven, Leuven, Belgium
| | - Ellen Deleus
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Matthias Lannoo
- Department of Abdominal Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium; Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Translational Research for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Jacobsen JM, Petersen N, Torz L, Gerstenberg MK, Pedersen K, Østergaard S, Wulff BS, Andersen B, Raun K, Christoffersen BØ, John LM, Reitman ML, Kuhre RE. Housing mice near vs. below thermoneutrality affects drug-induced weight loss but does not improve prediction of efficacy in humans. Cell Rep 2024; 43:114501. [PMID: 39067024 PMCID: PMC11380917 DOI: 10.1016/j.celrep.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Evaluation of weight loss drugs is usually performed in diet-induced obese mice housed at ∼22°C. This is a cold stress that increases energy expenditure by ∼35% compared to thermoneutrality (∼30°C), which may overestimate drug-induced weight loss. We investigated five anti-obesity mechanisms that have been in clinical development, comparing weight loss in mice housed at 22°C vs. 30°C. Glucagon-like peptide-1 (GLP-1), human fibroblast growth factor 21 (hFGF21), and melanocortin-4 receptor (MC4R) agonist induced similar weight losses. Peptide YY elicited greater vehicle-subtracted weight loss at 30°C (7.2% vs. 1.4%), whereas growth differentiation factor 15 (GDF15) was more effective at 22°C (13% vs. 6%). Independent of ambient temperature, GLP-1 and hFGF21 prevented the reduction in metabolic rate caused by weight loss. There was no simple rule for a better prediction of human drug efficacy based on ambient temperature, but since humans live at thermoneutrality, drug testing using mice should include experiments near thermoneutrality.
Collapse
Affiliation(s)
- Julie M Jacobsen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Natalia Petersen
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Lola Torz
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Kent Pedersen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Søren Østergaard
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte S Wulff
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte Andersen
- Diabetes, Obesity and NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Kirsten Raun
- Lead Portfolio Projects, Research and Early Development, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Linu M John
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Rune E Kuhre
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
8
|
Xie W, Sharma A, Kaushik H, Sharma L, Nistha, Anwer MK, Sachdeva M, Elossaily GM, Zhang Y, Pillappan R, Kaur M, Behl T, Shen B, Singla RK. Shaping the future of gastrointestinal cancers through metabolic interactions with host gut microbiota. Heliyon 2024; 10:e35336. [PMID: 39170494 PMCID: PMC11336605 DOI: 10.1016/j.heliyon.2024.e35336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/19/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Gastrointestinal (GI) cancers represent a significant global health challenge, driving relentless efforts to identify innovative diagnostic and therapeutic approaches. Recent strides in microbiome research have unveiled a previously underestimated dimension of cancer progression that revolves around the intricate metabolic interplay between GI cancers and the host's gut microbiota. This review aims to provide a comprehensive overview of these emerging metabolic interactions and their potential to catalyze a paradigm shift in precision diagnosis and therapeutic breakthroughs in GI cancers. The article underscores the groundbreaking impact of microbiome research on oncology by delving into the symbiotic connection between host metabolism and the gut microbiota. It offers valuable insights into tailoring treatment strategies to individual patients, thus moving beyond the traditional one-size-fits-all approach. This review also sheds light on novel diagnostic methodologies that could transform the early detection of GI cancers, potentially leading to more favorable patient outcomes. In conclusion, exploring the metabolic interactions between host gut microbiota and GI cancers showcases a promising frontier in the ongoing battle against these formidable diseases. By comprehending and harnessing the microbiome's influence, the future of precision diagnosis and therapeutic innovation for GI cancers appears more optimistic, opening doors to tailored treatments and enhanced diagnostic precision.
Collapse
Affiliation(s)
- Wen Xie
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Aditi Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P, 173229, India
| | - Hitesh Kaushik
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P, 173229, India
| | - Lalit Sharma
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P, 173229, India
| | - Nistha
- School of Pharmaceutical Sciences, Shoolini University, Solan, H.P, 173229, India
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Monika Sachdeva
- Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Gehan M. Elossaily
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh, 11597, Saudi Arabia
| | - Yingbo Zhang
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
| | - Ramkumar Pillappan
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Mangaluru, Karnataka, India
| | - Maninderjit Kaur
- Department of Pharmaceutical Sciences, lovely Professional University, Phagwara, India
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Sahibzada Ajit Singh Nagar, Punjab, India
| | - Bairong Shen
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rajeev K. Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 1444411, India
| |
Collapse
|
9
|
Vrabie AM, Totolici S, Delcea C, Badila E. Biomarkers in Heart Failure with Preserved Ejection Fraction: A Perpetually Evolving Frontier. J Clin Med 2024; 13:4627. [PMID: 39200768 PMCID: PMC11355893 DOI: 10.3390/jcm13164627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/28/2024] [Accepted: 08/05/2024] [Indexed: 09/02/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) represents a complex clinical syndrome, often very difficult to diagnose using the available tools. As the global burden of this disease is constantly growing, surpassing the prevalence of heart failure with reduced ejection fraction, during the last few years, efforts have focused on optimizing the diagnostic and prognostic pathways using an immense panel of circulating biomarkers. After the paradigm of HFpEF development emerged more than 10 years ago, suggesting the impact of multiple comorbidities on myocardial structure and function, several phenotypes of HFpEF have been characterized, with an attempt to find an ideal biomarker for each distinct pathophysiological pathway. Acknowledging the limitations of natriuretic peptides, hundreds of potential biomarkers have been evaluated, some of them demonstrating encouraging results. Among these, soluble suppression of tumorigenesis-2 reflecting myocardial remodeling, growth differentiation factor 15 as a marker of inflammation and albuminuria as a result of kidney dysfunction or, more recently, several circulating microRNAs have proved their incremental value. As the number of emerging biomarkers in HFpEF is rapidly expanding, in this review, we aim to explore the most promising available biomarkers linked to key pathophysiological mechanisms in HFpEF, outlining their utility for diagnosis, risk stratification and population screening, as well as their limitations.
Collapse
Affiliation(s)
- Ana-Maria Vrabie
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Stefan Totolici
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Caterina Delcea
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Elisabeta Badila
- Cardio-Thoracic Pathology Department, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (S.T.); (C.D.); (E.B.)
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
10
|
Zou T, Long Z, Wang S, Yao Q. Nausea and vomiting in pregnancy (NVP) in Chinese pregnant women: a cross-sectional study. BMC Pregnancy Childbirth 2024; 24:481. [PMID: 39014303 PMCID: PMC11251098 DOI: 10.1186/s12884-024-06686-7] [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: 10/19/2023] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND This study addresses the scarcity of research on nausea and vomiting in pregnancy (NVP) in China. It aims to explore the current NVP status in the country using validated questionnaires, analyze associated factors, and provide a useful reference for future research. The study also compares results from different assessment tools. METHODS Online questionnaires were utilized to gather data from 535 pregnant women across 24 provinces. Demographic, pregnancy, and NVP-related information were collected. NVP severity was assessed using Pregnancy-Unique Quantification of Emesis and Nausea (PUQE) and the Rhodes Index of Nausea, Vomiting, and Retching (RINVR) scales. Ordinal logistic regression identified factors linked to NVP severity. Differences between PUQE and RINVR assessments were compared. RESULTS NVP prevalence exceeded 90%, with 96.1% assessed by PUQE and 90.8% by RINVR. Incidence decreased from nausea to retching and vomiting. Severe NVP correlated with reduced gestational weight gain, younger age, fewer gestational weeks, and living in North (all P values < 0.05). There was moderate consistency between PUQE and RINVR assessments. The NVP prevalence assessed by the PUQE is higher than that assessed by the RINVR in the same population. However, the proportion of NVP levels above moderate assessed by RINVR is greater than that assessed by PUQE. CONCLUSIONS NVP is highly prevalent among Chinese pregnant women, with nausea being predominant. RINVR assessments may be better able to identify severe NVP, thereby improving the low treatment rates for severe NVP.
Collapse
Affiliation(s)
- Tong Zou
- West China Second University Hospital, Sichuan University, Chengdu, No. 20 Ren Min Nan Road, Sichuan, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | | | - Silu Wang
- Recovery Plus USA, New York, NY, 10019, USA
| | - Qiang Yao
- West China Second University Hospital, Sichuan University, Chengdu, No. 20 Ren Min Nan Road, Sichuan, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
11
|
Li J, Hu X, Xie Z, Li J, Huang C, Huang Y. Overview of growth differentiation factor 15 (GDF15) in metabolic diseases. Biomed Pharmacother 2024; 176:116809. [PMID: 38810400 DOI: 10.1016/j.biopha.2024.116809] [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: 03/18/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
GDF15 is a stress response cytokine and a distant member of the transforming growth factor beta (TGFβ) superfamily, its levels increase in response to cell stress and certain diseases in the serum. To exert its effects, GDF15 binds to glial-derived neurotrophic factor (GDNF) receptor alpha-like (GFRAL), which was firstly identified in 2017 and highly expressed in the brain stem. Many studies have demonstrated that elevated serum GDF15 is associated with anorexia and weight loss. Herein, we focus on the biology of GDF15, specifically how this circulating protein regulates appetite and metabolism in influencing energy homeostasis through its actions on hindbrain neurons to shed light on its impact on diseases such as obesity and anorexia/cachexia syndromes. It works as an endocrine factor and transmits metabolic signals leading to weight reduction effects by directly reducing appetite and indirectly affecting food intake through complex mechanisms, which could be a promising target for the treatment of energy-intake disorders.
Collapse
Affiliation(s)
- Jian Li
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, China
| | - Xiangjun Hu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zichuan Xie
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiajin Li
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Chen Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
12
|
Park MA, Whelan CJ, Ahmed S, Boeringer T, Brown J, Crowder SL, Gage K, Gregg C, Jeong DK, Jim HSL, Judge AR, Mason TM, Parker N, Pillai S, Qayyum A, Rajasekhara S, Rasool G, Tinsley SM, Schabath MB, Stewart P, West J, McDonald P, Permuth JB. Defining and Addressing Research Priorities in Cancer Cachexia through Transdisciplinary Collaboration. Cancers (Basel) 2024; 16:2364. [PMID: 39001427 PMCID: PMC11240731 DOI: 10.3390/cancers16132364] [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: 05/29/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
For many patients, the cancer continuum includes a syndrome known as cancer-associated cachexia (CAC), which encompasses the unintended loss of body weight and muscle mass, and is often associated with fat loss, decreased appetite, lower tolerance and poorer response to treatment, poor quality of life, and reduced survival. Unfortunately, there are no effective therapeutic interventions to completely reverse cancer cachexia and no FDA-approved pharmacologic agents; hence, new approaches are urgently needed. In May of 2022, researchers and clinicians from Moffitt Cancer Center held an inaugural retreat on CAC that aimed to review the state of the science, identify knowledge gaps and research priorities, and foster transdisciplinary collaborative research projects. This review summarizes research priorities that emerged from the retreat, examples of ongoing collaborations, and opportunities to move science forward. The highest priorities identified include the need to (1) evaluate patient-reported outcome (PRO) measures obtained in clinical practice and assess their use in improving CAC-related outcomes; (2) identify biomarkers (imaging, molecular, and/or behavioral) and novel analytic approaches to accurately predict the early onset of CAC and its progression; and (3) develop and test interventions (pharmacologic, nutritional, exercise-based, and through mathematical modeling) to prevent CAC progression and improve associated symptoms and outcomes.
Collapse
Affiliation(s)
- Margaret A. Park
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Christopher J. Whelan
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Sabeen Ahmed
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Tabitha Boeringer
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Joel Brown
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sylvia L. Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Kenneth Gage
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Christopher Gregg
- School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| | - Daniel K. Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Heather S. L. Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Andrew R. Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA;
| | - Tina M. Mason
- Department of Nursing Research, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nathan Parker
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Smitha Pillai
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Aliya Qayyum
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Sahana Rajasekhara
- Department of Supportive Care Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghulam Rasool
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Sara M. Tinsley
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Paul Stewart
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Jeffrey West
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Patricia McDonald
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Lexicon Pharmaceuticals, Inc., Woodlands, TX 77381, USA
| | - Jennifer B. Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| |
Collapse
|
13
|
Kwon YY, Hui S. IL-6 promotes tumor growth through immune evasion but is dispensable for cachexia. EMBO Rep 2024; 25:2592-2609. [PMID: 38671295 PMCID: PMC11169252 DOI: 10.1038/s44319-024-00144-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 03/26/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Various cytokines have been implicated in cancer cachexia. One such cytokine is IL-6, deemed as a key cachectic factor in mice inoculated with colon carcinoma 26 (C26) cells, a widely used cancer cachexia model. Here we tested the causal role of IL-6 in cancer cachexia by knocking out the IL-6 gene in C26 cells. We found that the growth of IL-6 KO tumors was dramatically delayed. More strikingly, while IL-6 KO tumors eventually reached the similar size as wild-type tumors, cachexia still took place, despite no elevation in circulating IL-6. In addition, the knockout of leukemia inhibitory factor (LIF), another IL-6 family cytokine proposed as a cachectic factor in the model, also affected tumor growth but not cachexia. We further showed an increase in the infiltration of immune cell population in the IL-6 KO tumors compared with wild-type controls and the defective IL-6 KO tumor growth was rescued in immunodeficient mice while cachexia was not. Thus, IL-6 promotes tumor growth by facilitating immune evasion but is dispensable for cachexia.
Collapse
Affiliation(s)
- Young-Yon Kwon
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
14
|
Sun Q, van de Lisdonk D, Ferrer M, Gegenhuber B, Wu M, Park Y, Tuveson DA, Tollkuhn J, Janowitz T, Li B. Area postrema neurons mediate interleukin-6 function in cancer cachexia. Nat Commun 2024; 15:4682. [PMID: 38824130 PMCID: PMC11144211 DOI: 10.1038/s41467-024-48971-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Interleukin-6 (IL-6) has been long considered a key player in cancer cachexia. It is believed that sustained elevation of IL-6 production during cancer progression causes brain dysfunctions, which ultimately result in cachexia. However, how peripheral IL-6 influences the brain remains poorly understood. Here we show that neurons in the area postrema (AP), a circumventricular structure in the hindbrain, is a critical mediator of IL-6 function in cancer cachexia in male mice. We find that circulating IL-6 can rapidly enter the AP and activate neurons in the AP and its associated network. Peripheral tumor, known to increase circulating IL-6, leads to elevated IL-6 in the AP, and causes potentiated excitatory synaptic transmission onto AP neurons and AP network hyperactivity. Remarkably, neutralization of IL-6 in the brain of tumor-bearing mice with an anti-IL-6 antibody attenuates cachexia and the hyperactivity in the AP network, and markedly prolongs lifespan. Furthermore, suppression of Il6ra, the gene encoding IL-6 receptor, specifically in AP neurons with CRISPR/dCas9 interference achieves similar effects. Silencing Gfral-expressing AP neurons also attenuates cancer cachectic phenotypes and AP network hyperactivity. Our study identifies a central mechanism underlying the function of peripheral IL-6, which may serve as a target for treating cancer cachexia.
Collapse
Affiliation(s)
- Qingtao Sun
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Daniëlle van de Lisdonk
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands
| | - Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Bruno Gegenhuber
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Melody Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Jessica Tollkuhn
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Bo Li
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, 310024, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, 310024, Zhejiang, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, 310024, Zhejiang, China.
| |
Collapse
|
15
|
Groarke JD, Crawford J, Collins SM, Lubaczewski SL, Breen DM, Harrington MA, Jacobs I, Qiu R, Revkin J, Rossulek MI, Saxena AR. Phase 2 study of the efficacy and safety of ponsegromab in patients with cancer cachexia: PROACC-1 study design. J Cachexia Sarcopenia Muscle 2024; 15:1054-1061. [PMID: 38500292 PMCID: PMC11154777 DOI: 10.1002/jcsm.13435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/06/2023] [Accepted: 12/27/2023] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Cancer cachexia is a multifactorial metabolic wasting syndrome characterized by anorexia, unintentional loss of weight involving both skeletal muscle and adipose tissues, progressive functional impairment and reduced survival. Therapeutic strategies for this serious condition are very limited. Growth differentiation factor 15 (GDF-15) is a cytokine that is implicated in cancer cachexia and may represent both a biomarker of cancer cachexia and a potential therapeutic target. Ponsegromab is a potent and selective humanized monoclonal antibody that inhibits GDF-15-mediated signalling. Preclinical and preliminary phase 1 data suggest that ponsegromab-mediated inactivation of circulating GDF-15 may lead to improvement in key characteristics of cachexia. The primary objective of this phase 2 study is to assess the effect of ponsegromab on body weight in patients with cancer, cachexia and elevated GDF-15 concentrations. Secondary objectives include assessing physical activity, physical function, actigraphy, appetite, nausea and vomiting, fatigue and safety. Exploratory objectives include evaluating pharmacokinetics, pharmacodynamics, immunogenicity, lumbar skeletal muscle index and Response Evaluation Criteria in Solid Tumors. METHODS Approximately 168 adults with non-small-cell lung, pancreatic or colorectal cancers who have cachexia and elevated GDF-15 concentrations will be randomized in a double-blind, placebo-controlled study (NCT05546476). Participants meeting eligibility criteria will be randomized 1:1:1:1 to one of three dose groups of ponsegromab (100, 200 or 400 mg) or matching placebo administered subcutaneously every 4 weeks for an initial 12-week treatment period. This is followed by optional open-label treatment with ponsegromab of 400 mg administered every 4 weeks for up to 1 year. The primary endpoint is mean change from baseline in body weight at Week 12. A mixed model for repeated measures followed by a Bayesian Emax model will be used for the primary analysis. Secondary endpoints include physical activity, physical function and actigraphy measured by remote digital sensors; patient-reported appetite-related symptoms assessed by Functional Assessment of Anorexia-Cachexia Therapy subscale scores; anorexia/appetite, nausea and vomiting, and fatigue evaluated according to questions from the Cancer-Related Cachexia Symptom Diary; and incidence of adverse events, safety laboratory tests, vital signs and electrocardiogram abnormalities. PERSPECTIVE Cancer-related cachexia is an area of significant unmet medical need. This study will support the clinical development of ponsegromab as a novel inhibitor of GDF-15, which may ameliorate key pathologies of cancer cachexia to improve patient symptoms, functionality and quality of life. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05546476.
Collapse
Affiliation(s)
| | | | - Susie M. Collins
- Global Biometrics and Data ManagementPfizer R&D UK LtdSandwichKentUK
| | - Shannon L. Lubaczewski
- Early Clinical Development and Biomedicine Artificial IntelligencePfizer IncCollegevillePAUSA
| | | | | | - Ira Jacobs
- Global Product DevelopmentPfizer IncNew YorkNYUSA
| | - Ruolun Qiu
- Clinical PharmacologyPfizer IncCambridgeMAUSA
| | - James Revkin
- Internal Medicine Research UnitPfizer IncCambridgeMAUSA
| | | | | |
Collapse
|
16
|
Park H, Lee CH. The contribution of the nervous system in the cancer progression. BMB Rep 2024; 57:167-175. [PMID: 38523371 PMCID: PMC11058356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/09/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
Cancer progression is driven by genetic mutations, environmental factors, and intricate interactions within the tumor microenvironment (TME). The TME comprises of diverse cell types, such as cancer cells, immune cells, stromal cells, and neuronal cells. These cells mutually influence each other through various factors, including cytokines, vascular perfusion, and matrix stiffness. In the initial or developmental stage of cancer, neurotrophic factors such as nerve growth factor, brain-derived neurotrophic factor, and glial cell line-derived neurotrophic factor are associated with poor prognosis of various cancers by communicating with cancer cells, immune cells, and peripheral nerves within the TME. Over the past decade, research has been conducted to prevent cancer growth by controlling the activation of neurotrophic factors within tumors, exhibiting a novel attemt in cancer treatment with promising results. More recently, research focusing on controlling cancer growth through regulation of the autonomic nervous system, including the sympathetic and parasympathetic nervous systems, has gained significant attention. Sympathetic signaling predominantly promotes tumor progression, while the role of parasympathetic signaling varies among different cancer types. Neurotransmitters released from these signalings can directly or indirectly affect tumor cells or immune cells within the TME. Additionally, sensory nerve significantly promotes cancer progression. In the advanced stage of cancer, cancer-associated cachexia occurs, characterized by tissue wasting and reduced quality of life. This process involves the pathways via brainstem growth and differentiation factor 15-glial cell line-derived neurotrophic factor receptor alpha-like signaling and hypothalamic proopiomelanocortin neurons. Our review highlights the critical role of neurotrophic factors as well as central nervous system on the progression of cancer, offering promising avenues for targeted therapeutic strategies. [BMB Reports 2024; 57(4): 167-175].
Collapse
Affiliation(s)
- Hongryeol Park
- Department of Tissue Morphogenesis, Max-Planck Institute for Molecular Biomedicine, Muenster D-48149, Germany, Chuncheon 24252, Korea
| | - Chan Hee Lee
- Department of Biomedical Science, Hallym University, Chuncheon 24252, Korea
- Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
17
|
Crawford J, Calle RA, Collins SM, Weng Y, Lubaczewski SL, Buckeridge C, Wang EQ, Harrington MA, Tarachandani A, Rossulek MI, Revkin JH. A Phase Ib First-In-Patient Study Assessing the Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of Ponsegromab in Participants with Cancer and Cachexia. Clin Cancer Res 2024; 30:489-497. [PMID: 37982848 PMCID: PMC10831332 DOI: 10.1158/1078-0432.ccr-23-1631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/18/2023] [Accepted: 11/16/2023] [Indexed: 11/21/2023]
Abstract
PURPOSE Cachexia is common in patients with advanced cancer and is associated with elevated serum growth differentiation factor 15 (GDF-15) concentrations. This first-in-patient (phase Ib), 24-week study assessed use of ponsegromab, a mAb against GDF-15, in adults with advanced cancer, cachexia, and elevated GDF-15 serum concentration. PATIENTS AND METHODS Participants (n = 10) received open-label ponsegromab subcutaneous 200 mg every 3 weeks for 12 weeks in addition to standard-of-care anticancer treatment. Ponsegromab safety, tolerability, and pharmacokinetics were assessed in addition to serum GDF-15 concentrations and exploratory measures of efficacy. RESULTS No treatment-related treatment-emergent adverse events, injection site reactions, or adverse trends in clinical laboratory tests, vital signs, or electrocardiogram parameters attributable to ponsegromab were identified. Median serum unbound GDF-15 concentration at baseline was 2.269 ng/mL. Following initiation of study treatment, median unbound GDF-15 concentrations were below the lower limit of quantification (0.0424 ng/mL) from day 1 (3 hours postdose) through week 15. Increases in body weight were observed at all time points during the treatment and follow-up periods. A least-squares mean (SE) increase of 4.63 (1.98) kg was observed at week 12, an increase of approximately 6.6% relative to baseline. Ponsegromab-mediated improvements in actigraphy-based assessments of physical activity and in quality of life, including appetite as assessed by Functional Assessment of Anorexia-Cachexia Therapy total and subscale scores, were also observed. CONCLUSIONS Ponsegromab was well tolerated, suppressed serum GDF-15 concentrations, and demonstrated preliminary evidence of efficacy. These findings support the continued development of ponsegromab for the treatment of cachexia.
Collapse
Affiliation(s)
- Jeffrey Crawford
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | - Roberto A. Calle
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Susie M. Collins
- Global Biometrics and Data Management, Pfizer R&D UK Ltd, Sandwich, Kent, United Kingdom
| | - Yan Weng
- Clinical Pharmacology, Pfizer Inc, Cambridge, Massachusetts
| | - Shannon L. Lubaczewski
- Early Clinical Development Biomedicine Artificial Intelligence, Pfizer Inc, Collegeville, Pennsylvania
| | - Clare Buckeridge
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Ellen Q. Wang
- Clinical Pharmacology, Pfizer Inc, New York, New York
| | | | | | | | - James H. Revkin
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| |
Collapse
|
18
|
Chen YC, Chen JH, Tsai CF, Wu CT, Chang PC, Yeh WL. Inhibition of tumor migration and invasion by fenofibrate via suppressing epithelial-mesenchymal transition in breast cancers. Toxicol Appl Pharmacol 2024; 483:116818. [PMID: 38215994 DOI: 10.1016/j.taap.2024.116818] [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: 09/12/2023] [Revised: 12/16/2023] [Accepted: 01/09/2024] [Indexed: 01/14/2024]
Abstract
The recurrence and metastasis in breast cancer within 3 years after the chemotherapies or surgery leads to poor prognosis with approximately 1-year overall survival. Large-scale scanning research studies have shown that taking lipid-lowering drugs may assist to reduce the risk of death from many cancers, since cholesterol in lipid rafts are essential for maintain integral membrane structure and functional signaling regulation. In this study, we examined five lipid-lowering drugs: swertiamarin, gemfibrozil, clofibrate, bezafibrate, and fenofibrate in triple-negative breast cancer, which is the most migration-prone subtype. Using human and murine triple-negative breast cancer cell lines (Hs 578 t and 4 T1), we found that fenofibrate displays the highest potential in inhibiting the colony formation, wound healing, and transwell migration. We further discovered that fenofibrate reduces the activity of pro-metastatic enzymes, matrix metalloproteinases (MMP)-9 and MMP-2. In addition, epithelial markers including E-cadherin and Zonula occludens-1 are increased, whereas mesenchymal markers including Snail, Twist and α-smooth muscle actin are attenuated. Furthermore, we found that fenofibrate downregulates ubiquitin-dependent GDF-15 degradation, which leads to enhanced GDF-15 expression that inhibits cell migration. Besides, nuclear translocation of FOXO1 is also upregulated by fenofibrate, which may responsible for GDF-15 expression. In summary, fenofibrate with anti-cancer ability hinders TNBC from migration and invasion, and may be beneficial to repurposing use of fenofibrate.
Collapse
Affiliation(s)
- Yen-Chang Chen
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung 404333, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 88, Sec. 1, Fengxing Road, Taichung 427213, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, No.500 Lioufeng Road, Taichung 413305, Taiwan
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, No. 2, Yude Road, Taichung 404332, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics and Medical Engineering, Asia University, No.500 Lioufeng Road, Taichung 413305, Taiwan
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung 404333, Taiwan; Department of Biochemistry, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung 404333, Taiwan.
| |
Collapse
|
19
|
Neshan M, Tsilimigras DI, Han X, Zhu H, Pawlik TM. Molecular Mechanisms of Cachexia: A Review. Cells 2024; 13:252. [PMID: 38334644 PMCID: PMC10854699 DOI: 10.3390/cells13030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/18/2024] [Accepted: 01/28/2024] [Indexed: 02/10/2024] Open
Abstract
Cachexia is a condition characterized by substantial loss of body weight resulting from the depletion of skeletal muscle and adipose tissue. A considerable fraction of patients with advanced cancer, particularly those who have been diagnosed with pancreatic or gastric cancer, lung cancer, prostate cancer, colon cancer, breast cancer, or leukemias, are impacted by this condition. This syndrome manifests at all stages of cancer and is associated with an unfavorable prognosis. It heightens the susceptibility to surgical complications, chemotherapy toxicity, functional impairments, breathing difficulties, and fatigue. The early detection of patients with cancer cachexia has the potential to enhance both their quality of life and overall survival rates. Regarding this matter, blood biomarkers, although helpful, possess certain limitations and do not exhibit universal application. Additionally, the available treatment options for cachexia are currently limited, and there is a lack of comprehensive understanding of the underlying molecular pathways associated with this condition. Thus, this review aims to provide an overview of molecular mechanisms associated with cachexia and potential therapeutic targets for the development of effective treatments for this devastating condition.
Collapse
Affiliation(s)
- Mahdi Neshan
- Department of General Surgery, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd 8915887857, Iran;
| | - Diamantis I. Tsilimigras
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Xu Han
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Hua Zhu
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA; (D.I.T.); (X.H.); (H.Z.)
| |
Collapse
|
20
|
Li X, Guan H, Ma C, Dai Y, Su J, Chen X, Yuan Q, Wang J. Combination of bulk RNA sequencing and scRNA sequencing uncover the molecular characteristics of MAPK signaling in kidney renal clear cell carcinoma. Aging (Albany NY) 2024; 16:1414-1439. [PMID: 38217548 PMCID: PMC10866414 DOI: 10.18632/aging.205436] [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: 07/11/2023] [Accepted: 12/01/2023] [Indexed: 01/15/2024]
Abstract
The MAPK signaling pathway significantly impacts cancer progression and resistance; however, its functions remain incompletely assessed across various cancers, particularly in kidney renal clear cell carcinoma (KIRC). Therefore, there is an urgent need for comprehensive pan-cancer investigations of MAPK signaling, particularly within the context of KIRC. In this research, we obtained TCGA pan-cancer multi-omics data and conducted a comprehensive analysis of the genomic and transcriptomic characteristics of the MAPK signaling pathway. For in-depth investigation in KIRC, status of MAPK pathway was quantitatively estimated by ssGSEA and Ward algorithm was utilized for cluster analysis. Molecular characteristics and clinical prognoses of KIRC patients with distinct MAPK activities were comprehensively explored using a series of bioinformatics algorithms. Subsequently, a combination of LASSO and COX regression analyses were utilized sequentially to construct a MAPK-related signature to help identify the risk level of each sample. Patients in the C1 subtype exhibited relatively higher levels of MAPK signaling activity, which were associated with abundant immune cell infiltration and favorable clinical outcomes. Single-cell RNA sequencing (scRNA-seq) analysis of KIRC samples identified seven distinct cell types, and endothelial cells in tumor tissues had obviously higher MAPK scores than normal tissues. The immunohistochemistry results indicated the reduced expression levels of PAPSS1, MAP3K11, and SPRED1 in KIRC samples. In conclusion, our study represents the first integration of bulk RNA sequencing and single-cell RNA sequencing to elucidate the molecular characteristics of MAPK signaling in KIRC, providing a solid foundation for precision oncology.
Collapse
Affiliation(s)
- Xiunan Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hewen Guan
- Department of Dermatology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Chuanyu Ma
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yunfei Dai
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ji Su
- Department of Urology, Central Hospital of Benxi, Benxi, Liaoning, China
| | - Xu Chen
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Qihang Yuan
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jianbo Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
21
|
Wang Y, Dong Z, An Z, Jin W. Cancer cachexia: Focus on cachexia factors and inter-organ communication. Chin Med J (Engl) 2024; 137:44-62. [PMID: 37968131 PMCID: PMC10766315 DOI: 10.1097/cm9.0000000000002846] [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: 05/25/2023] [Indexed: 11/17/2023] Open
Abstract
ABSTRACT Cancer cachexia is a multi-organ syndrome and closely related to changes in signal communication between organs, which is mediated by cancer cachexia factors. Cancer cachexia factors, being the general name of inflammatory factors, circulating proteins, metabolites, and microRNA secreted by tumor or host cells, play a role in secretory or other organs and mediate complex signal communication between organs during cancer cachexia. Cancer cachexia factors are also a potential target for the diagnosis and treatment. The pathogenesis of cachexia is unclear and no clear effective treatment is available. Thus, the treatment of cancer cachexia from the perspective of the tumor ecosystem rather than from the perspective of a single molecule and a single organ is urgently needed. From the point of signal communication between organs mediated by cancer cachexia factors, finding a deeper understanding of the pathogenesis, diagnosis, and treatment of cancer cachexia is of great significance to improve the level of diagnosis and treatment. This review begins with cancer cachexia factors released during the interaction between tumor and host cells, and provides a comprehensive summary of the pathogenesis, diagnosis, and treatment for cancer cachexia, along with a particular sight on multi-organ signal communication mediated by cancer cachexia factors. This summary aims to deepen medical community's understanding of cancer cachexia and may conduce to the discovery of new diagnostic and therapeutic targets for cancer cachexia.
Collapse
Affiliation(s)
- Yongfei Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Zikai Dong
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Ziyi An
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| | - Weilin Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, China
| |
Collapse
|
22
|
Hughes AE, Freathy RM. Nausea and vomiting in pregnancy linked to hormone from fetus. Nature 2024; 625:670-671. [PMID: 38093044 DOI: 10.1038/d41586-023-03940-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
|
23
|
Snoke DB, Bellefleur E, Rehman HT, Carson JA, Poynter ME, Dittus KL, Toth MJ. Skeletal muscle adaptations in patients with lung cancer: Longitudinal observations from the whole body to cellular level. J Cachexia Sarcopenia Muscle 2023; 14:2579-2590. [PMID: 37727010 PMCID: PMC10751417 DOI: 10.1002/jcsm.13332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Cancer and its treatment can adversely affect skeletal muscle, impacting physical function, treatment response and survival. No studies, however, have comprehensively characterized these muscle adaptations longitudinally in human patients at the cellular level. METHODS We examined skeletal muscle size and function from the whole body to the sub-cellular level in 11 patients with non-small cell lung cancer (NSCLC; 6 male/5 female, mean age 58 ± 3 years) studied over a 2-month observation period starting during their first cycle of standard of care cancer treatment and in 11 age- and sex-matched healthy controls (HC) without a current or past history of cancer. Biopsies of the vastus lateralis were performed to assess muscle fibre size, contractility and mitochondrial content, along with assessments of physical function, whole muscle size and function, and circulating cytokines. RESULTS Body weight, composition and thigh muscle area and density were unaltered over time in patients with NSCLC, while muscle density was lower in patients with NSCLC versus HC (P = 0.03). Skeletal muscle fibre size decreased by 18% over time in patients (all P = 0.02) and was lower than HC (P = 0.02). Mitochondrial fractional area and density did not change over time in patients, but fractional area was lower in patients with NSCLC compared with HC (subsarcolemmal, P = 0.04; intermyofibrillar, P = 0.03). Patients with NSCLC had higher plasma concentrations of IL-6 (HC 1.40 ± 0.50; NSCLC 4.71 ± 4.22; P < 0.01), GDF-15 (HC 569 ± 166; NSCLC 2071 ± 1168; P < 0.01) and IL-8/CXCL8 (HC 4.9 ± 1.8; NSCLC 10.1 ± 6.0; P = 0.02) compared with HC, but there were no changes in inflammatory markers in patients with NSCLC over time. No changes were observed in markers of satellite cell activation or DNA damage in patients and no group differences were noted with HC. Whole-muscle strength was preserved over time in patients with NSCLC coincident with improved single fibre contractility. CONCLUSIONS This study is the first to comprehensively examine longitudinal alterations in skeletal muscle fibre size and function in patients with NSCLC and suggests that muscle fibre atrophy occurs during cancer treatment despite weight stability and no changes in conventional clinical measurements of whole body or thigh muscle size over this period.
Collapse
Affiliation(s)
- Deena B. Snoke
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Emma Bellefleur
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Hibba Tul Rehman
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
- University of Vermont Cancer CenterUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - James A. Carson
- Department of Physical TherapyThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Matthew E. Poynter
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Kim L. Dittus
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
- University of Vermont Cancer CenterUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| | - Michael J. Toth
- Department of MedicineUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
- University of Vermont Cancer CenterUniversity of Vermont Larner College of MedicineBurlingtonVermontUSA
| |
Collapse
|
24
|
Callaway CS, Mouchantat LM, Bitler BG, Bonetto A. Mechanisms of Ovarian Cancer-Associated Cachexia. Endocrinology 2023; 165:bqad176. [PMID: 37980602 PMCID: PMC10699881 DOI: 10.1210/endocr/bqad176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/02/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
Cancer-associated cachexia occurs in 50% to 80% of cancer patients and is responsible for 20% to 30% of cancer-related deaths. Cachexia limits survival and treatment outcomes, and is a major contributor to morbidity and mortality during cancer. Ovarian cancer is one of the leading causes of cancer-related deaths in women, and recent studies have begun to highlight the prevalence and clinical impact of cachexia in this population. Here, we review the existing understanding of cachexia pathophysiology and summarize relevant studies assessing ovarian cancer-associated cachexia in clinical and preclinical studies. In clinical studies, there is increased evidence that reduced skeletal muscle mass and quality associate with worse outcomes in subjects with ovarian cancer. Mouse models of ovarian cancer display cachexia, often characterized by muscle and fat wasting alongside inflammation, although they remain underexplored relative to other cachexia-associated cancer types. Certain soluble factors have been identified and successfully targeted in these models, providing novel therapeutic targets for mitigating cachexia during ovarian cancer. However, given the relatively low number of studies, the translational relevance of these findings is yet to be determined and requires more research. Overall, our current understanding of ovarian cancer-associated cachexia is insufficient and this review highlights the need for future research specifically aimed at exploring mechanisms of ovarian cancer-associated cachexia by using unbiased approaches and animal models representative of the clinical landscape of ovarian cancer.
Collapse
Affiliation(s)
- Chandler S Callaway
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Lila M Mouchantat
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Benjamin G Bitler
- Department of Obstetrics & Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
25
|
Ling T, Zhang J, Ding F, Ma L. Role of growth differentiation factor 15 in cancer cachexia (Review). Oncol Lett 2023; 26:462. [PMID: 37780545 PMCID: PMC10534279 DOI: 10.3892/ol.2023.14049] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023] Open
Abstract
Growth differentiation factor 15 (GDF15), a member of the transforming growth factor-β family, is a stress-induced cytokine. Under normal circumstances, the expression of GDF15 is low in most tissues. It is highly expressed during tissue injury, inflammation, oxidative stress and cancer. GDF15 has been established as a biomarker in patients with cancer, and is associated with cancer cachexia (CC) and poor survival. CC is a multifactorial metabolic disorder characterized by severe muscle and adipose tissue atrophy, loss of appetite, anemia and bone loss. Cachexia leads to reductions in quality of life and tolerance to anticancer therapy, and results in a poor prognosis in cancer patients. Dysregulated GDF15 levels have been discovered in patients with CC and animal models, where they have been found to be involved in anorexia and weight loss. Although studies have suggested that GDF15 mediates anorexia and weight loss in CC through its neuroreceptor, glial cell-lineage neurotrophic factor family receptor α-like, the effects of GDF15 on CC and the potential regulatory mechanisms require further elucidation. In the present review, the characteristics of GDF15 and its roles and molecular mechanisms in CC are elaborated. The targeting of GDF15 as a potential therapeutic strategy for CC is also discussed.
Collapse
Affiliation(s)
- Tingting Ling
- Department of Oncology, Affiliated Hospital of Weifang Medical College, Weifang, Shandong 261000, P.R. China
| | - Jing Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical College, Weifang, Shandong 261000, P.R. China
| | - Fuwan Ding
- Department of Endocrinology, Yancheng Third People's Hospital, Yancheng, Jiangsu 224001, P.R. China
| | - Lanlan Ma
- Graduate School, Weifang Medical College, Weifang, Shandong 261000, P.R. China
| |
Collapse
|
26
|
Alexopoulou F, Buch-Månson N, Pedersen SL, Vrang N, Fink LN, Strømgaard K. Identification of GDF15 peptide fragments inhibiting GFRAL receptor signaling. Peptides 2023; 168:171063. [PMID: 37495041 DOI: 10.1016/j.peptides.2023.171063] [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: 06/20/2023] [Accepted: 07/10/2023] [Indexed: 07/28/2023]
Abstract
Growth differentiation factor 15 (GDF15) is believed to be a major causative factor for cancer-induced cachexia. Recent elucidation of the central circuits involved in GDF15 function and its signaling through the glial cell-derived neurotrophic factor family receptor α-like (GFRAL) has prompted the interest of targeting the GDF15-GFRAL signaling for energy homeostasis and body weight regulation. Here, we applied advanced peptide technologies to identify GDF15 peptide fragments inhibiting GFRAL signaling. SPOT peptide arrays revealed binding of GDF15 C-terminal peptide fragments to the extracellular domain of GFRAL. Parallel solid-phase peptide synthesis allowed for generation of complementary GDF15 peptide libraries and their subsequent functional evaluation in cells expressing the GFRAL/RET receptor complex. We identified a series of C-terminal fragments of GDF15 inhibiting GFRAL activity in the micromolar range. These novel GFRAL peptide inhibitors could serve as valuable tools for further development of peptide therapeutics towards the treatment of cachexia and other wasting disorders.
Collapse
Affiliation(s)
- Flora Alexopoulou
- Gubra Aps, Hørsholm, DK-2970 Hørsholm, Denmark; Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | | | | | - Niels Vrang
- Gubra Aps, Hørsholm, DK-2970 Hørsholm, Denmark
| | | | - Kristian Strømgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
27
|
Wang J, Tan S, Gianotti L, Wu G. Evaluation and management of body composition changes in cancer patients. Nutrition 2023; 114:112132. [PMID: 37441827 DOI: 10.1016/j.nut.2023.112132] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 06/06/2023] [Accepted: 06/10/2023] [Indexed: 07/15/2023]
Abstract
Wasting in cancer patients has long been recognized as a condition that adversely affects cancer patients' quality of life, treatment tolerance, and oncological outcomes. Historically, this condition was mainly evaluated by changes in body weight. However, this approach is not quite accurate because body weight is the overall change of all body compartments. Conditions such as edema and ascites can mask the severity of muscle and adipose tissue depletion. Changes in body composition assessment in cancer patients have historically been underappreciated because of the limited availability of measurement tools. As more evidence highlighting the importance of body composition has emerged, it is imperative to apply a more precise evaluation of nutritional status and a more targeted approach to provide nutritional support for cancer patients. In this review, we will discuss the modalities for evaluating body composition and how to manage body composition changes in cancer patients.
Collapse
Affiliation(s)
- Junjie Wang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shanjun Tan
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Luca Gianotti
- School of Medicine and Surgery, University of Milano-Bicocca, and HBP Surgery Unit, and Foundation IRCCS San Gerardo, Monza, Italy.
| | - Guohao Wu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Winter LM, Reinhardt D, Schatter A, Tissen V, Wiora H, Gerlach D, Tontsch-Grunt U, Colbatzky F, Stierstorfer B, Yun SW. Molecular basis of GDF15 induction and suppression by drugs in cardiomyocytes and cancer cells toward precision medicine. Sci Rep 2023; 13:12061. [PMID: 37495707 PMCID: PMC10372009 DOI: 10.1038/s41598-023-38450-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/08/2023] [Indexed: 07/28/2023] Open
Abstract
GDF15 has recently emerged as a key driver of the development of various disease conditions including cancer cachexia. Not only the tumor itself but also adverse effects of chemotherapy have been reported to contribute to increased GDF15. Although regulation of GDF15 transcription by BET domain has recently been reported, the molecular mechanisms of GDF15 gene regulation by drugs are still unknown, leaving uncertainty about the safe and effective therapeutic strategies targeting GDF15. We screened various cardiotoxic drugs and BET inhibitors for their effects on GDF15 regulation in human cardiomyocytes and cancer cell lines and analyzed in-house and public gene signature databases. We found that DNA damaging drugs induce GDF15 in cardiomyocytes more strongly than drugs with other modes of action. In cancer cells, GDF15 induction varied depending on drug- and cell type-specific gene signatures including mutations in PI3KCA, TP53, BRAF and MUC16. GDF15 suppression by BET inhibition is particularly effective in cancer cells with low activity of the PI3K/Akt axis and high extracellular concentrations of pantothenate. Our findings provide insights that the risk for GDF15 overexpression and concomitant cachexia can be reduced by a personalized selection of anticancer drugs and patients for precision medicine.
Collapse
Affiliation(s)
- Lisa-Maria Winter
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Diana Reinhardt
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Ariane Schatter
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Vivien Tissen
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Heike Wiora
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Daniel Gerlach
- Boehringer Ingelheim RCV, GmbH & Co KG, 1120, Vienna, Austria
| | | | - Florian Colbatzky
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Birgit Stierstorfer
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany
| | - Seong-Wook Yun
- Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Strasse 65, 88397, Biberach an Der Riß, Germany.
| |
Collapse
|
29
|
Kerr HL, Krumm K, Lee I(I, Anderson B, Christiani A, Strait L, Breckheimer BA, Irwin B, Jiang A(S, Rybachok A, Chen A, Caeiro L, Dacek E, Hall DB, Kostyla CH, Hales LM, Soliman TM, Garcia JM. EXT418, a novel long-acting ghrelin, mitigates Lewis lung carcinoma induced cachexia in mice. J Cachexia Sarcopenia Muscle 2023; 14:1337-1348. [PMID: 36942661 PMCID: PMC10235874 DOI: 10.1002/jcsm.13211] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND Ghrelin is a potential therapy for cachexia due to its orexigenic properties and anabolic effects on muscle and fat. However, its clinical use is limited by the short half-life of active (acylated) ghrelin (~11 min in humans). EXT418 is a novel long-acting, constitutively active ghrelin analog created by covalently linking it to a vitamin D derivative. Here, we evaluated the effects and mechanisms of action of EXT418 on Lewis lung carcinoma (LLC)-induced cachexia in mice. METHODS Male C57BL/6J mice (5- to 7-month-old) were implanted with 1 × 106 heat-killed (HK) or live LLC cells. When the tumour was palpable, mice were injected with vehicle (T + V) or EXT418 daily (T + 418 Daily, 0.25 mg/kg/day) or every other day (T + 418 EOD, 0.5 mg/kg/EOD) for up to 14 days, whereas HK-treated mice were given vehicle (HK + V). Subsets of T + 418 Daily or EOD-treated mice were pair-fed to the T + V group. Body composition and grip strength were evaluated before tumour implantation and at the end of the experiment. Molecular markers were probed in muscles upon termination. RESULTS In tumour-bearing mice, administration of EXT418 daily or EOD partially prevented weight loss (T + V vs. T + 418 Daily, P = 0.030; and vs. T + 418 EOD, P = 0.020). Similar effects were observed in whole body fat and lean body mass. Grip strength in tumour-bearing mice was improved by EXT418 daily (P = 0.010) or EOD (P = 0.008) administration compared with vehicle-treated mice. These effects of EXT418 on weight and grip strength were partially independent of food intake. EXT418 daily administration also improved type IIA (P = 0.015), IIB (P = 0.037) and IIX (P = 0.050) fibre cross-sectional area (CSA) in tibialis anterior (TA) and EXT418 EOD improved CSA of IIB fibres in red gastrocnemius (GAS; P = 0.005). In skeletal muscles, tumour-induced increases in atrogenes Fbxo32 and Trim63 were ameliorated by EXT418 treatments (TA and GAS/plantaris, PL), which were independent of food intake. EXT418 administration decreased expression of the mitophagy marker Bnip3 (GAS/PL; P ≤ 0.010). Similar effects of EXT418 EOD were observed in p62 (GAS/PL; P = 0.039). In addition, EXT418 treatments ameliorated the tumour-induced elevation in muscle Il6 transcript levels (TA and GAS/PL), independently of food intake. Il-6 transcript levels in adipose tissue and circulating IL-10 were elevated in response to the tumour but these increases were not significant with EXT418 administration. Tumour mass was not altered by EXT418. CONCLUSIONS EXT418 mitigates LLC-induced cachexia by attenuating skeletal muscle inflammation, proteolysis, and mitophagy, without affecting tumour mass and partially independent of food intake.
Collapse
Affiliation(s)
- Haiming L. Kerr
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Kora Krumm
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Ian (In‐gi) Lee
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Barbara Anderson
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Anthony Christiani
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Lena Strait
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Beatrice A. Breckheimer
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Brynn Irwin
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Alice (Siyi) Jiang
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Artur Rybachok
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Amanda Chen
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Lucas Caeiro
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | - Elizabeth Dacek
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| | | | - Caroline H. Kostyla
- Extend Biosciences, Inc.St. NewtonMassachusettsUSA
- Present address:
Atalanta TherapeuticsBostonMassachusettsUSA
| | | | | | - Jose M. Garcia
- Geriatric Research, Education and Clinical CenterVeterans Affairs Puget Sound Health Care SystemSeattleWashingtonUSA
- Department of Medicine, Division of Gerontology and Geriatric MedicineUniversity of Washington School of MedicineWashingtonSeattleUSA
| |
Collapse
|
30
|
Martin A, Gallot YS, Freyssenet D. Molecular mechanisms of cancer cachexia-related loss of skeletal muscle mass: data analysis from preclinical and clinical studies. J Cachexia Sarcopenia Muscle 2023; 14:1150-1167. [PMID: 36864755 PMCID: PMC10235899 DOI: 10.1002/jcsm.13073] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 06/15/2022] [Accepted: 08/14/2022] [Indexed: 03/04/2023] Open
Abstract
Cancer cachexia is a systemic hypoanabolic and catabolic syndrome that diminishes the quality of life of cancer patients, decreases the efficiency of therapeutic strategies and ultimately contributes to decrease their lifespan. The depletion of skeletal muscle compartment, which represents the primary site of protein loss during cancer cachexia, is of very poor prognostic in cancer patients. In this review, we provide an extensive and comparative analysis of the molecular mechanisms involved in the regulation of skeletal muscle mass in human cachectic cancer patients and in animal models of cancer cachexia. We summarize data from preclinical and clinical studies investigating how the protein turnover is regulated in cachectic skeletal muscle and question to what extent the transcriptional and translational capacities, as well as the proteolytic capacity (ubiquitin-proteasome system, autophagy-lysosome system and calpains) of skeletal muscle are involved in the cachectic syndrome in human and animals. We also wonder how regulatory mechanisms such as insulin/IGF1-AKT-mTOR pathway, endoplasmic reticulum stress and unfolded protein response, oxidative stress, inflammation (cytokines and downstream IL1ß/TNFα-NF-κB and IL6-JAK-STAT3 pathways), TGF-ß signalling pathways (myostatin/activin A-SMAD2/3 and BMP-SMAD1/5/8 pathways), as well as glucocorticoid signalling, modulate skeletal muscle proteostasis in cachectic cancer patients and animals. Finally, a brief description of the effects of various therapeutic strategies in preclinical models is also provided. Differences in the molecular and biochemical responses of skeletal muscle to cancer cachexia between human and animals (protein turnover rates, regulation of ubiquitin-proteasome system and myostatin/activin A-SMAD2/3 signalling pathways) are highlighted and discussed. Identifying the various and intertwined mechanisms that are deregulated during cancer cachexia and understanding why they are decontrolled will provide therapeutic targets for the treatment of skeletal muscle wasting in cancer patients.
Collapse
Affiliation(s)
- Agnès Martin
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| | - Yann S. Gallot
- LBEPS, Univ Evry, IRBA, Université Paris SaclayEvryFrance
| | - Damien Freyssenet
- Laboratoire Interuniversitaire de Biologie de la Motricité EA 7424, Univ LyonUniversité Jean Monnet Saint‐EtienneSaint‐Priest‐en‐JarezFrance
| |
Collapse
|
31
|
Jaschke NP, Rachner TD. Cancer cachexia as a blueprint for treating obesity. Trends Endocrinol Metab 2023:S1043-2760(23)00087-5. [PMID: 37173233 DOI: 10.1016/j.tem.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023]
Abstract
Effective pharmacological treatments to achieve significant and sustained weight loss in obese individuals remain limited. Here, we apply a 'reverse engineering' approach to cancer cachexia, an extreme form of dysregulated energy balance resulting in net catabolism. We discuss three phenotypic features of the disease, summarize the underlying molecular checkpoints, and explore their translation to obesity research. We then provide examples for established pharmaceuticals, which follow a reverse engineering logic, and propose additional targets that may be of relevance for future studies. Finally, we argue that approaching diseases from this perspective may prove useful as a generic strategy to fuel the development of innovative therapies.
Collapse
Affiliation(s)
- Nikolai P Jaschke
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden, Dresden, Germany.
| | - Tilman D Rachner
- Division of Endocrinology, Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
32
|
Al-Sawaf O, Weiss J, Skrzypski M, Lam JM, Karasaki T, Zambrana F, Kidd AC, Frankell AM, Watkins TBK, Martínez-Ruiz C, Puttick C, Black JRM, Huebner A, Bakir MA, Sokač M, Collins S, Veeriah S, Magno N, Naceur-Lombardelli C, Prymas P, Toncheva A, Ward S, Jayanth N, Salgado R, Bridge CP, Christiani DC, Mak RH, Bay C, Rosenthal M, Sattar N, Welsh P, Liu Y, Perrimon N, Popuri K, Beg MF, McGranahan N, Hackshaw A, Breen DM, O'Rahilly S, Birkbak NJ, Aerts HJWL, Jamal-Hanjani M, Swanton C. Body composition and lung cancer-associated cachexia in TRACERx. Nat Med 2023; 29:846-858. [PMID: 37045997 PMCID: PMC7614477 DOI: 10.1038/s41591-023-02232-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/24/2023] [Indexed: 04/14/2023]
Abstract
Cancer-associated cachexia (CAC) is a major contributor to morbidity and mortality in individuals with non-small cell lung cancer. Key features of CAC include alterations in body composition and body weight. Here, we explore the association between body composition and body weight with survival and delineate potential biological processes and mediators that contribute to the development of CAC. Computed tomography-based body composition analysis of 651 individuals in the TRACERx (TRAcking non-small cell lung Cancer Evolution through therapy (Rx)) study suggested that individuals in the bottom 20th percentile of the distribution of skeletal muscle or adipose tissue area at the time of lung cancer diagnosis, had significantly shorter lung cancer-specific survival and overall survival. This finding was validated in 420 individuals in the independent Boston Lung Cancer Study. Individuals classified as having developed CAC according to one or more features at relapse encompassing loss of adipose or muscle tissue, or body mass index-adjusted weight loss were found to have distinct tumor genomic and transcriptomic profiles compared with individuals who did not develop such features. Primary non-small cell lung cancers from individuals who developed CAC were characterized by enrichment of inflammatory signaling and epithelial-mesenchymal transitional pathways, and differentially expressed genes upregulated in these tumors included cancer-testis antigen MAGEA6 and matrix metalloproteinases, such as ADAMTS3. In an exploratory proteomic analysis of circulating putative mediators of cachexia performed in a subset of 110 individuals from TRACERx, a significant association between circulating GDF15 and loss of body weight, skeletal muscle and adipose tissue was identified at relapse, supporting the potential therapeutic relevance of targeting GDF15 in the management of CAC.
Collapse
Affiliation(s)
- Othman Al-Sawaf
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Jakob Weiss
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Diagnostic and Interventional Radiology, University Freiburg, Freiburg, Germany
| | - Marcin Skrzypski
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | | | - Andrew C Kidd
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Thomas B K Watkins
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Clare Puttick
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Mateo Sokač
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Susie Collins
- Early Clinical Development, Pfizer UK Ltd, Cambridge, UK
| | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Neil Magno
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | - Paulina Prymas
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Antonia Toncheva
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Sophia Ward
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Nick Jayanth
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Roberto Salgado
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - David C Christiani
- Department of Medicine, Massachusetts General Hospital/Harvard Medicine School, and Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Raymond H Mak
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Camden Bay
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Michael Rosenthal
- Department of Radiology, Brigham and Women's Hospital and Dana-Farber Cancer Institute, and Harvard Medical School, Boston, MA, USA
| | - Naveed Sattar
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Paul Welsh
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ying Liu
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, USA
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, St. John's, Newfoundland and Labrador, Burnaby, Canada
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, British Colombia, Canada
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Danna M Breen
- Internal Medicine Research Unit, Pfizer, Cambridge, MA, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Nicolai J Birkbak
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Bioinformatics Research Centre, Aarhus University, Aarhus, Denmark
| | - Hugo J W L Aerts
- Artificial Intelligence in Medicine (AIM) Program, Mass General Brigham, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Radiology and Nuclear Medicine, CARIM & GROW, Maastricht University, Maastricht, The Netherlands
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
33
|
Saroul N, Tardif N, Pereira B, Dissard A, Montrieul L, Sanchez P, Salles J, Petersen JE, Jakobson T, Gilain L, Mom T, Boirie Y, Rooyakers O, Walrand S. Conditioned Media from Head and Neck Cancer Cell Lines and Serum Samples from Head and Neck Cancer Patients Drive Catabolic Pathways in Cultured Muscle Cells. Cancers (Basel) 2023; 15:cancers15061843. [PMID: 36980729 PMCID: PMC10047086 DOI: 10.3390/cancers15061843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The role of secreted factors from the tumor cells in driving cancer cachexia and especially muscle loss is unknown. We wanted to study both the action of secreted factors from head and neck cancer (HNC) cell lines and circulating factors in HNC patients on skeletal muscle protein catabolism. METHODS Conditioned media (CM) made from head and neck cancer cell lines and mix of sera from head and neck cancer (HNC) patients were incubated for 48 h with human myotubes. The atrophy and the catabolic pathway were monitored in myotubes. The patients were classified regarding their skeletal muscle loss observed at the outset of management. RESULTS Tumor CM (TCM) was able to produce atrophy on myotubes as compared with control CM (CCM). However, a mix of sera from HNC patients was not able to produce atrophy in myotubes. Despite this discrepancy on atrophy, we observed a similar regulation of the catabolic pathways by the tumor-conditioned media and mix of sera from cancer patients. The catabolic response after incubation with the mix of sera seemed to depend on the muscle loss seen in patients. CONCLUSION This study found evidence that the atrophy observed in HNC patients cannot be solely explained by a deficit in food intake.
Collapse
Affiliation(s)
- Nicolas Saroul
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Biostatistics Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Nicolas Tardif
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Huddinge, Sweden
| | - Bruno Pereira
- Biostatistics Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Alexis Dissard
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Laura Montrieul
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Phelipe Sanchez
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Jérôme Salles
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Jens Erik Petersen
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
| | - Towe Jakobson
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
| | - Laurent Gilain
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Thierry Mom
- Otolaryngology Head and Neck Surgery Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Yves Boirie
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Clinical Nutrition Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| | - Olav Rooyakers
- Anesthesiology and Intensive Care, Department of Clinical Science Intervention and Technology, CLINTEC, Karolinska Institutet, 141 86 Huddinge, Sweden
- Division of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 171 77 Huddinge, Sweden
| | - Stéphane Walrand
- Human Nutrition Unit, INRAE, Auvergne Human Nutrition Research Center, Clermont Auvergne University, CHU de Clermont-Ferrand France, INRAE, UNH, 63000 Clermont-Ferrand, France
- Clinical Nutrition Department, CHU de Clermont-Ferrand France, 63000 Clermont-Ferrand, France
| |
Collapse
|
34
|
Morita-Tanaka S, Yamada T, Takayama K. The landscape of cancer cachexia in advanced non-small cell lung cancer: a narrative review. Transl Lung Cancer Res 2023; 12:168-180. [PMID: 36762058 PMCID: PMC9903087 DOI: 10.21037/tlcr-22-561] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023]
Abstract
Background and Objective Cancer cachexia presents with weight loss, anorexia, and fatigue and worsens the prognosis and quality of life of cancer patients. We aimed to summarize the current relevant discourse in the literature about cancer cachexia in the setting of non-small cell lung carcinoma and the possible current and future treatments. Methods We conduct a narrative review of the literature on the landscape of cancer cachexia in the context of non-small cell lung cancer, multimodality therapy, markers, imaging, tumor biology, pathology, chemoprevention, and technical advances. Key Content and Findings The need for appropriate intervention for cancer cachexia is increasing as the prognosis of patients with advanced non-small cell lung cancer is improving with advances in treatment. Tumor cells play a role in the pathogenesis of cachexia, where they release factors that elicit the production of inflammatory cytokines by the immune system resulting in decreased appetite, abnormal energy metabolism, and skeletal muscle degeneration. Comorbid chronic lung diseases are associated with pulmonary cachexia and sarcopenia and commonly occur in the context of lung cancer, further contributing to the increased incidence of cachexia in patients with lung cancer. Currently, a ghrelin-like agonist, anamorelin, is approved for the treatment of cancer cachexia and is used in clinical practice in Japan. The role that nutritional and exercise therapies can play as added treatments must be further explored. Conclusions Cancer cachexia remains a poorly understood phenomenon, and awareness must be raised through educational activities for health care providers and patient family members. In addition, new therapeutics targeting cancer cachexia, such as GDF-15 antibodies, are in development, and further progress is expected.
Collapse
Affiliation(s)
- Satomi Morita-Tanaka
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tadaaki Yamada
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koichi Takayama
- Department of Pulmonary Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
35
|
Kim-Muller JY, Song L, LaCarubba Paulhus B, Pashos E, Li X, Rinaldi A, Joaquim S, Stansfield JC, Zhang J, Robertson A, Pang J, Opsahl A, Boucher M, Breen D, Hales K, Sheikh A, Wu Z, Zhang BB. GDF15 neutralization restores muscle function and physical performance in a mouse model of cancer cachexia. Cell Rep 2023; 42:111947. [PMID: 36640326 DOI: 10.1016/j.celrep.2022.111947] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/06/2022] [Accepted: 12/16/2022] [Indexed: 01/11/2023] Open
Abstract
Cancer cachexia is a disorder characterized by involuntary weight loss and impaired physical performance. Decline in physical performance of patients with cachexia is associated with poor quality of life, and currently there are no effective pharmacological interventions that restore physical performance. Here we examine the effect of GDF15 neutralization in a mouse model of cancer-induced cachexia (TOV21G) that manifests weight loss and muscle function impairments. With comprehensive assessments, our results demonstrate that cachectic mice treated with the anti-GDF15 antibody mAB2 exhibit body weight gain with near-complete restoration of muscle mass and markedly improved muscle function and physical performance. Mechanistically, the improvements induced by GDF15 neutralization are primarily attributed to increased caloric intake, while altered gene expression in cachectic muscles is restored in caloric-intake-dependent and -independent manners. The findings indicate potential of GDF15 neutralization as an effective therapy to enhance physical performance of patients with cachexia.
Collapse
Affiliation(s)
- Ja Young Kim-Muller
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - LouJin Song
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Brianna LaCarubba Paulhus
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Evanthia Pashos
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Xiangping Li
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Anthony Rinaldi
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Stephanie Joaquim
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - John C Stansfield
- Biostatistics, Early Clinical Development, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Jiangwei Zhang
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 10777 Science Center Dr., San Diego, CA, USA
| | - Andrew Robertson
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Jincheng Pang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Alan Opsahl
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer Worldwide Research, Development & Medical, 445 Eastern Point Rd., Groton, CT, USA
| | - Danna Breen
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Abdul Sheikh
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA
| | - Bei B Zhang
- Internal Medicine Research Unit, Pfizer Worldwide Research, Development & Medical, 1 Portland St., Cambridge, MA, USA.
| |
Collapse
|
36
|
Sun Q, van de Lisdonk D, Ferrer M, Gegenhuber B, Wu M, Tollkuhn J, Janowitz T, Li B. Area postrema neurons mediate interleukin-6 function in cancer-associated cachexia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523716. [PMID: 36711916 PMCID: PMC9882141 DOI: 10.1101/2023.01.12.523716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Interleukin-6 (IL-6) has been long considered a key player in cancer-associated cachexia 1-15 . It is believed that sustained elevation of IL-6 production during cancer progression causes brain dysfunctions, which ultimately result in cachexia 16-20 . However, how peripheral IL-6 influences the brain remains poorly understood. Here we show that neurons in the area postrema (AP), a circumventricular structure in the hindbrain, mediate the function of IL-6 in cancer-associated cachexia in mice. We found that circulating IL-6 can rapidly enter the AP and activate AP neurons. Peripheral tumor, known to increase circulating IL-6 1-5,15,18,21-23 , leads to elevated IL-6 and neuronal hyperactivity in the AP, and causes potentiated excitatory synaptic transmission onto AP neurons. Remarkably, neutralization of IL-6 in the brain of tumor-bearing mice with an IL-6 antibody prevents cachexia, reduces the hyperactivity in an AP network, and markedly prolongs lifespan. Furthermore, suppression of Il6ra , the gene encoding IL-6 receptor, specifically in AP neurons with CRISPR/dCas9 interference achieves similar effects. Silencing of Gfral-expressing AP neurons also ameliorates the cancer-associated cachectic phenotypes and AP network hyperactivity. Our study identifies a central mechanism underlying the function of peripheral IL-6, which may serve as a target for treating cancer-associated cachexia.
Collapse
|
37
|
Wu K, Liu X, Meng X, Cao L, Li H, Bi Y, Wang M, Wang M, Jiang Y. Sauchinone alleviates dextran sulfate sodium-induced ulcerative colitis via NAD(P)H dehydrogenase [quinone] 1/NF-kB pathway and gut microbiota. Front Microbiol 2023; 13:1084257. [PMID: 36699607 PMCID: PMC9868758 DOI: 10.3389/fmicb.2022.1084257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023] Open
Abstract
Objective This study evaluated the effects of sauchinone on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mice model and investigated the underlying mechanisms of the downstream pathway and gut microbiota. Methods The UC mice model was induced by DSS. The disease phenotypes were determined through pathological symptoms (body weight and disease activity index score), inflammation markers (histological and inflammatory factor detections), and colonic mucosal barrier damage (detection of tight junction proteins). The level of the NF-κB pathway was detected through marker proteins. Database and bioinformatics analyses were used to predict sauchinone-mediated downstream molecules that were previously identified by expression analysis. Mouse feces were collected to detect the V3-V4 region of the 16S rRNA gene. Results In DSS-induced UC mice, sauchinone alleviated pathological symptoms, inhibited inflammation, and prevented mucosal barrier damage. Sauchinone further inhibited the NF-κB pathway by upregulating NAD (P) H dehydrogenase [quinone] 1 (NQO1) in DSS-induced UC mice. Moreover, sauchinone regulated the diversity and composition of the gut microbiota in mice, stimulating the growth of Firmicutes and inhibiting the growth of Proteobacteria and Bacteroidetes. Conclusion Therefore, sauchinone exerted therapeutic effects on UC in mice by regulating the NQO1/NF-κB pathway and altering the gut microbiota. This provides a theoretical basis for developing sauchinone as a therapeutic agent and extends our understanding of its bioactivity.
Collapse
Affiliation(s)
- Kun Wu
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xianjun Liu
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Xianglong Meng
- Department of Burns Surgery, The First Hospital of Jilin University, Changchun, China
| | - Lingling Cao
- School of Clinical Medical, Changchun University of Chinese Medicine, Changchun, China
| | - Hao Li
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Yingxin Bi
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Mengyuan Wang
- College of Biological and Food Engineering, Jilin Engineering Normal University, Changchun, China
| | - Mingchuan Wang
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Jiang
- Department of Gastrointestinal Colorectal and Anal Surgery, The China-Japan Union Hospital of Jilin University, Changchun, China,*Correspondence: Yang Jiang, ✉
| |
Collapse
|
38
|
Wang Y, An Z, Lin D, Jin W. Targeting cancer cachexia: Molecular mechanisms and clinical study. MedComm (Beijing) 2022; 3:e164. [PMID: 36105371 PMCID: PMC9464063 DOI: 10.1002/mco2.164] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022] Open
Abstract
Cancer cachexia is a complex systemic catabolism syndrome characterized by muscle wasting. It affects multiple distant organs and their crosstalk with cancer constitute cancer cachexia environment. During the occurrence and progression of cancer cachexia, interactions of aberrant organs with cancer cells or other organs in a cancer cachexia environment initiate a cascade of stress reactions and destroy multiple organs including the liver, heart, pancreas, intestine, brain, bone, and spleen in metabolism, neural, and immune homeostasis. The role of involved organs turned from inhibiting tumor growth into promoting cancer cachexia in cancer progression. In this review, we depicted the complicated relationship of cancer cachexia with the metabolism, neural, and immune homeostasis imbalance in multiple organs in a cancer cachexia environment and summarized the treatment progress in recent years. And we discussed the molecular mechanism and clinical study of cancer cachexia from the perspective of multiple organs metabolic, neurological, and immunological abnormalities. Updated understanding of cancer cachexia might facilitate the exploration of biomarkers and novel therapeutic targets of cancer cachexia.
Collapse
Affiliation(s)
- Yong‐Fei Wang
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Zi‐Yi An
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Dong‐Hai Lin
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| | - Wei‐Lin Jin
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
39
|
De Paepe B. The Cytokine Growth Differentiation Factor-15 and Skeletal Muscle Health: Portrait of an Emerging Widely Applicable Disease Biomarker. Int J Mol Sci 2022; 23:ijms232113180. [PMID: 36361969 PMCID: PMC9654287 DOI: 10.3390/ijms232113180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/21/2022] [Accepted: 10/26/2022] [Indexed: 12/04/2022] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a stress-induced transforming growth factor-β superfamily cytokine with versatile functions in human health. Elevated GDF-15 blood levels associate with multiple pathological conditions, and are currently extensively explored for diagnosis, and as a means to monitor disease progression and evaluate therapeutic responses. This review analyzes GDF-15 in human conditions specifically focusing on its association with muscle manifestations of sarcopenia, mitochondrial myopathy, and autoimmune and viral myositis. The use of GDF-15 as a widely applicable health biomarker to monitor muscle disease is discussed, and its potential as a therapeutic target is explored.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
40
|
Ferrara M, Samaden M, Ruggieri E, Vénéreau E. Cancer cachexia as a multiorgan failure: Reconstruction of the crime scene. Front Cell Dev Biol 2022; 10:960341. [PMID: 36158184 PMCID: PMC9493094 DOI: 10.3389/fcell.2022.960341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cachexia is a devastating syndrome associated with the end-stage of several diseases, including cancer, and characterized by body weight loss and severe muscle and adipose tissue wasting. Although different cancer types are affected to diverse extents by cachexia, about 80% of all cancer patients experience this comorbidity, which highly reduces quality of life and response to therapy, and worsens prognosis, accounting for more than 25% of all cancer deaths. Cachexia represents an urgent medical need because, despite several molecular mechanisms have been identified, no effective therapy is currently available for this devastating syndrome. Most studies focus on skeletal muscle, which is indeed the main affected and clinically relevant organ, but cancer cachexia is characterized by a multiorgan failure. In this review, we focus on the current knowledge on the multiple tissues affected by cachexia and on the biomarkers with the attempt to define a chronological pathway, which might be useful for the early identification of patients who will undergo cachexia. Indeed, it is likely that the inefficiency of current therapies might be attributed, at least in part, to their administration in patients at the late stages of cachexia.
Collapse
Affiliation(s)
- Michele Ferrara
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Samaden
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Ruggieri
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emilie Vénéreau
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
41
|
Transforming Growth Factor-Beta Signaling in Cancer-Induced Cachexia: From Molecular Pathways to the Clinics. Cells 2022; 11:cells11172671. [PMID: 36078078 PMCID: PMC9454487 DOI: 10.3390/cells11172671] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 02/06/2023] Open
Abstract
Cachexia is a metabolic syndrome consisting of massive loss of muscle mass and function that has a severe impact on the quality of life and survival of cancer patients. Up to 20% of lung cancer patients and up to 80% of pancreatic cancer patients are diagnosed with cachexia, leading to death in 20% of them. The main drivers of cachexia are cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), macrophage inhibitory cytokine 1 (MIC-1/GDF15) and transforming growth factor-beta (TGF-β). Besides its double-edged role as a tumor suppressor and activator, TGF-β causes muscle loss through myostatin-based signaling, involved in the reduction in protein synthesis and enhanced protein degradation. Additionally, TGF-β induces inhibin and activin, causing weight loss and muscle depletion, while MIC-1/GDF15, a member of the TGF-β superfamily, leads to anorexia and so, indirectly, to muscle wasting, acting on the hypothalamus center. Against this background, the blockade of TGF-β is tested as a potential mechanism to revert cachexia, and antibodies against TGF-β reduced weight and muscle loss in murine models of pancreatic cancer. This article reviews the role of the TGF-β pathway and to a minor extent of other molecules including microRNA in cancer onset and progression with a special focus on their involvement in cachexia, to enlighten whether TGF-β and such other players could be potential targets for therapy.
Collapse
|
42
|
Queiroz AL, Dantas E, Ramsamooj S, Murthy A, Ahmed M, Zunica ERM, Liang RJ, Murphy J, Holman CD, Bare CJ, Ghahramani G, Wu Z, Cohen DE, Kirwan JP, Cantley LC, Axelrod CL, Goncalves MD. Blocking ActRIIB and restoring appetite reverses cachexia and improves survival in mice with lung cancer. Nat Commun 2022; 13:4633. [PMID: 35941104 PMCID: PMC9360437 DOI: 10.1038/s41467-022-32135-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/13/2022] [Indexed: 12/30/2022] Open
Abstract
Cancer cachexia is a common, debilitating condition with limited therapeutic options. Using an established mouse model of lung cancer, we find that cachexia is characterized by reduced food intake, spontaneous activity, and energy expenditure accompanied by muscle metabolic dysfunction and atrophy. We identify Activin A as a purported driver of cachexia and treat with ActRIIB-Fc, a decoy ligand for TGF-β/activin family members, together with anamorelin (Ana), a ghrelin receptor agonist, to reverse muscle dysfunction and anorexia, respectively. Ana effectively increases food intake but only the combination of drugs increases lean mass, restores spontaneous activity, and improves overall survival. These beneficial effects are limited to female mice and are dependent on ovarian function. In agreement, high expression of Activin A in human lung adenocarcinoma correlates with unfavorable prognosis only in female patients, despite similar expression levels in both sexes. This study suggests that multimodal, sex-specific, therapies are needed to reverse cachexia.
Collapse
Affiliation(s)
- Andre Lima Queiroz
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shakti Ramsamooj
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Anirudh Murthy
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Mujmmail Ahmed
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Roger J Liang
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jessica Murphy
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
- Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Corey D Holman
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Curtis J Bare
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Gregory Ghahramani
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Zhidan Wu
- Internal Medicine Research Unit, Pfizer Global R&D, Cambridge, MA, USA
| | - David E Cohen
- Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - John P Kirwan
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
43
|
Wang Y, Chen J, Chen C, Peng H, Lin X, Zhao Q, Chen S, Wang X. Growth differentiation factor-15 overexpression promotes cell proliferation and predicts poor prognosis in cerebral lower-grade gliomas correlated with hypoxia and glycolysis signature. Life Sci 2022; 302:120645. [PMID: 35588865 DOI: 10.1016/j.lfs.2022.120645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/15/2022] [Accepted: 05/12/2022] [Indexed: 12/30/2022]
Abstract
AIMS Growth differentiation factor-15 (GDF15) plays complex and controversial roles in cancer. In this study, the prognostic value and the exact biological function of GDF15 in cerebral lower-grade gliomas (LGGs) and its potential molecular targets were examined. MAIN METHODS Wilcoxon signed-rank test and logistic regression were applied to analyze associations between GDF15 expression and clinical characteristics using the Cancer Genome Atlas (TCGA) database. Overall survival was analyzed using Kaplan-Meier and Cox analyses. Gene set enrichment analysis (GSEA) and the hypoxia risk model was conducted to identify the potential molecular mechanisms underlying the effects of GDF15 on LGGs tumorigenesis. The biological function of GDF15 was examined using gain- and loss-of-function experiments, and a recombinant hGDF15 protein in LGG SW1783 cells in vitro. KEY FINDINGS We found that higher GDF15 expression is associated with poor clinical features in LGG patients, and an independent risk factor for overall survival among LGG patients. GSEA results showed that the poor prognostic role of GDF15 in LGGs is related to hypoxia and glycolysis signatures, which was further validated using the hypoxia risk model. Furthermore, GDF15 overexpression facilitated cell proliferation, while GDF15 siRNA inhibits cell proliferation in LGG SW1783 cells. In addition, GDF15 was upregulated upon CoCl2 treatment which induces hypoxia, correlating with the upregulation of the expressions of HIF-1α and glycolysis-related key genes in SW1783 cells. SIGNIFICANCE GDF15 may promote LGG tumorigenesis that is associated with the hypoxia and glycolysis pathways, and thus could serve as a promising molecular target for LGG prevention and therapy.
Collapse
Affiliation(s)
- Ying Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jiajun Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chaojie Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - He Peng
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xiaojian Lin
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Qian Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Shengjia Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xingya Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, PR China.
| |
Collapse
|
44
|
Timur B, Guney G. The role of serum ADAMTS-1 levels in Hyperemesis Gravidarum. BMC Pregnancy Childbirth 2022; 22:499. [PMID: 35725415 PMCID: PMC9208199 DOI: 10.1186/s12884-022-04832-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/10/2022] [Indexed: 11/19/2022] Open
Abstract
Background We aimed to investigate the levels of ADAMTS-1, which is secreted from the extracellular matrix during trophoblastic invasion in hyperemesis gravidarum (HEG). Methods In this cross-sectional study, we compared 45 HEG patients aged between 21 and 34 in terms of ADAMTS-1 levels with a control group consisting of 44 healthy pregnant women. The demographic characteristics and several laboratory parameters of the patients were recorded. Both groups were also compared in terms of ketonuria. We evaluated the correlation between ADAMTS-1 levels and ketonuria. Results The 2 groups were matched in terms of age, gestational age, gravidity, parity, and body mass index. Some inflammatory markers, such as neutrophil count, MPV, PDW, and PCT levels, were significantly higher in the HEG groups compared to the control group (all p < 0.05). However, mean MCV and serum TSH levels were statistically significantly lower in this group (both p < 0.001). ADAMTS-1 levels were 12.6 ± 1.4 ng/ml in the HEG group and 6.2 ± 1.6 ng/ml in the control group (p < 0.001). It was significantly and positively correlated with urine ketone, neutrophil count, and PDW, whereas negatively correlated with MCV and TSH value in the HEG group. ROC analysis showed that a threshold value of 11.275 ng/ml for ADAMTS-1 predicted HEG patients with a sensitivity of 60% and specificity of 95.5%. Conclusion ADAMTS-1 serum levels are increased in HEG patients, and there is a positive correlation between ADAMTS-1 levels and ketonuria.
Collapse
Affiliation(s)
- Burcu Timur
- Department of Obstetrics and Gynecology, Ordu University Training and Research Hospital, Bucak District, Nefsi Bucak Street, Ordu, 52200, Turkey.
| | - Gurhan Guney
- Department of Obstetrics and Gynecology, Balikesir University Medical Faculty, Balıkesir, Turkey
| |
Collapse
|
45
|
Wiggs MP, Beaudry AG, Law ML. Cardiac Remodeling in Cancer-Induced Cachexia: Functional, Structural, and Metabolic Contributors. Cells 2022; 11:cells11121931. [PMID: 35741060 PMCID: PMC9221803 DOI: 10.3390/cells11121931] [Citation(s) in RCA: 6] [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: 04/29/2022] [Revised: 06/01/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer cachexia is a syndrome of progressive weight loss and muscle wasting occurring in many advanced cancer patients. Cachexia significantly impairs quality of life and increases mortality. Cardiac atrophy and dysfunction have been observed in patients with cachexia, which may contribute to cachexia pathophysiology. However, relative to skeletal muscle, little research has been carried out to understand the mechanisms of cardiomyopathy in cachexia. Here, we review what is known clinically about the cardiac changes occurring in cachexia, followed by further discussion of underlying physiological and molecular mechanisms contributing to cachexia-induced cardiomyopathy. Impaired cardiac contractility and relaxation may be explained by a complex interplay of significant heart muscle atrophy and metabolic remodeling, including mitochondrial dysfunction. Because cardiac muscle has fundamental differences compared to skeletal muscle, understanding cardiac-specific effects of cachexia may bring light to unique therapeutic targets and ultimately improve clinical management for patients with cancer cachexia.
Collapse
Affiliation(s)
- Michael P. Wiggs
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA; (M.P.W.); (A.G.B.)
| | - Anna G. Beaudry
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA; (M.P.W.); (A.G.B.)
| | - Michelle L. Law
- Department of Human Sciences and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA
- Correspondence: ; Tel.: +1-(254)-710-6003
| |
Collapse
|
46
|
Cancer Cachexia and Antitumor Immunity: Common Mediators and Potential Targets for New Therapies. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060880. [PMID: 35743911 PMCID: PMC9225288 DOI: 10.3390/life12060880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 12/23/2022]
Abstract
Cancer cachexia syndrome (CCS) is a multifactorial metabolic syndrome affecting a significant proportion of patients. CCS is characterized by progressive weight loss, alterations of body composition and a systemic inflammatory status, which exerts a major impact on the host’s innate and adaptive immunity. Over the last few years, the development of immune checkpoint inhibitors (ICIs) transformed the treatment landscape for a wide spectrum of malignancies, creating an unprecedented opportunity for long term remissions in a significant subset of patients. Early clinical data indicate that CCS adversely impairs treatment outcomes of patients receiving ICIs. We herein reviewed existing evidence on the potential links between the mechanisms that promote the catabolic state in CCS and those that impair the antitumor immune response. We show that the biological mediators and processes leading to the development of CCS may also participate in the modulation and the sustainment of an immune suppressive tumor microenvironment and impaired anti-tumor immunity. Moreover, we demonstrate that the deregulation of the host’s metabolic homeostasis in cancer cachexia is associated with resistance to ICIs. Further research on the interrelation between cancer cachexia and anti-tumor immunity is required for the effective management of resistance to immunotherapy in this specific but large subgroup of ICI treated individuals.
Collapse
|
47
|
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
|
48
|
Hegde M, Daimary UD, Girisa S, Kumar A, Kunnumakkara AB. Tumor cell anabolism and host tissue catabolism-energetic inefficiency during cancer cachexia. Exp Biol Med (Maywood) 2022; 247:713-733. [PMID: 35521962 DOI: 10.1177/15353702221087962] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cancer-associated cachexia (CC) is a pathological condition characterized by sarcopenia, adipose tissue depletion, and progressive weight loss. CC is driven by multiple factors such as anorexia, excessive catabolism, elevated energy expenditure by growing tumor mass, and inflammatory mediators released by cancer cells and surrounding tissues. In addition, endocrine system, systemic metabolism, and central nervous system (CNS) perturbations in combination with cachexia mediators elicit exponential elevation in catabolism and reduced anabolism in skeletal muscle, adipose tissue, and cardiac muscle. At the molecular level, mechanisms of CC include inflammation, reduced protein synthesis, and lipogenesis, elevated proteolysis and lipolysis along with aggravated toxicity and complications of chemotherapy. Furthermore, CC is remarkably associated with intolerance to anti-neoplastic therapy, poor prognosis, and increased mortality with no established standard therapy. In this context, we discuss the spatio-temporal changes occurring in the various stages of CC and highlight the imbalance of host metabolism. We provide how multiple factors such as proteasomal pathways, inflammatory mediators, lipid and protein catabolism, glucocorticoids, and in-depth mechanisms of interplay between inflammatory molecules and CNS can trigger and amplify the cachectic processes. Finally, we highlight current diagnostic approaches and promising therapeutic interventions for CC.
Collapse
Affiliation(s)
- Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India.,DBT-AIST International Center for Translational and Environmental Research, Indian Institute of Technology-Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
49
|
Talbert EE, Guttridge DC. Emerging signaling mediators in the anorexia-cachexia syndrome of cancer. Trends Cancer 2022; 8:397-403. [PMID: 35190301 PMCID: PMC9035074 DOI: 10.1016/j.trecan.2022.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/11/2022]
Abstract
The cachexia syndrome in cancer is characterized by weight loss resulting from the combination of anorexia and atrophy of adipose and skeletal muscle. For decades, inflammatory circulatory factors have been identified to regulate wasting, but inhibitors of these factors have not yielded the same clinical benefit as in animal models. Therefore, additional mediators of cachexia likely regulate this syndrome, and such factors might be more suitable for targeted intervention. We highlight several anorexia-cachexia signaling mediators, including activin A, myostatin, GDF15, and lipocalin-2. We discuss current evidence that these factors associate with cachexia in cancer patients, and summarize translational efforts including essential early-phase clinical trials. We conclude with thoughts on targeted and personalized approaches for future anti-cachexia treatments.
Collapse
Affiliation(s)
- Erin E Talbert
- Department of Health and Human Physiology, and the Holden Comprehensive Cancer Center, University Iowa, Iowa City, IA 52242, USA
| | - Denis C Guttridge
- Department of Pediatrics, Darby Children's Research Institute, and the Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
50
|
Zhang W, Sun W, Gu X, Miao C, Feng L, Shen Q, Liu X, Zhang X. GDF-15 in tumor-derived exosomes promotes muscle atrophy via Bcl-2/caspase-3 pathway. Cell Death Dis 2022; 8:162. [PMID: 35379793 PMCID: PMC8980041 DOI: 10.1038/s41420-022-00972-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/04/2022] [Accepted: 03/23/2022] [Indexed: 12/22/2022]
Abstract
Tumor-derived exosomes are emerging mediators of cancer cachexia, a kind of multifactorial syndrome characterized by serious loss of skeletal muscle mass and function. Our previous study had showed that microRNAs in exosomes of C26 colon tumor cells were involved in induction of muscle atrophy. Here, we focus on studying proteins in tumor-derived exosomes which might also contribute to the development of cancer cachexia. Results of comparing the protein profiles of cachexic C26 exosomes and non-cachexic MC38 exosomes suggested that growth differentiation factor 15 (GDF-15) was rich in C26 exosomes. Western blotting analysis confirmed the higher levels of GDF-15 in C26 cells and C26 exosomes, compared with that of MC38 cells. Results of animal study also showed that GDF-15 was rich in tumor tissues, serum exosomes, and gastrocnemius (GA) muscle tissues of C26 tumor-bearing mice. GDF-15 protein could directly induce muscle atrophy of cultured C2C12 myotubes via regulating Bcl-2/caspase-3 pathways. What’s more, overexpression of GDF-15 in MC38 cells could increase the potency of MC38 conditioned medium or exosomes in inducing muscle atrophy. Knockdown of GDF-15 in C26 cells decreased the potency of C26 conditioned medium or exosomes in inducing muscle atrophy. These results suggested that GDF-15 in tumor-derived exosomes could contribute to induction of muscle atrophy and also supported the possibility of targeting GDF-15 in treatment of cancer cachexia.
Collapse
Affiliation(s)
- Wanli Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Weikuan Sun
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xiaofan Gu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Chunxiao Miao
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Lixing Feng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Qiang Shen
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China
| | - Xuan Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiongwen Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China.
| |
Collapse
|