Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 897
Filter
1.
Clin Nutr ; 43(7): 1800-1808, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38861892

ABSTRACT

BACKGROUND & AIMS: Our study aims to determine whether myostatin (MSTN) is associated with muscle mass and strength in individuals with cancer or obesity, as well as with cancer cachexia (CC) or sarcopenic obesity (SO). METHODS: The ACTICA study included individuals with CC (n = 70) or without CC (NC, n = 73). The MYDIASECRET study included individuals with obesity evaluated before (T0) and 3 months (T3) after bariatric surgery (n = 62). Body composition was assessed using bioelectrical impedance analysis (BIA). Skeletal muscle mass (SMM) and appendicular SMM (ASMM) were calculated from Janssen's and Sergi's equations, respectively, and expressed as indexes (SMMI and ASMMI). Handgrip strength (HGS) was assessed using a Jamar hand-held dynamometer. MSTN plasma levels were measured using ELISA. Spearman's coefficient was used to correlate MSTN with muscle mass and strength. Receiver operating characteristic (ROC) curve analysis was performed to identify an optimal MSTN cutoff level for the prediction of CC or SO. RESULTS: In the ACTICA study, muscle mass and strength were lower in CC individuals than in NC individuals (SMMI: 8.0 kg/m2vs 9.0 kg/m2, p = 0.004; ASMMI: 6.2 kg/m2vs 7.2 kg/m2, p < 0.001; HGS: 28 kg vs 38 kg, p < 0.001). MSTN was also lower in CC individuals than in NC individuals (1434 pg/mL vs 2149 pg/mL, p < 0.001). Muscle mass and strength were positively correlated with MSTN (SMMI: R = 0.500, p < 0.001; ASMMI: R = 0.479, p < 0.001; HGS: R = 0.495, p < 0.001). ROC curve analysis showed a MSTN cutoff level of 1548 pg/mL (AUC 0.684, sensitivity 57%, specificity 75%, p < 0.001) for the prediction of CC. In the MYDIASECRET study, muscle mass and strength were reduced at T3 (SMMI: -8%, p < 0.001; ASMMI: -12%, p < 0.001; HGS: -6%, p = 0.005). MSTN was also reduced at T3 (1773 pg/mL vs 2582 pg/mL, p < 0.001). Muscle mass and strength were positively correlated with MSTN at T0 and T3 (SMMI-T0: R = 0.388, p = 0.002; SMMI-T3: R = 0.435, p < 0.001; HGS-T0: R = 0.337, p = 0.007; HGS-T3: R = 0.313, p = 0.013). ROC curve analysis showed a MSTN cutoff level of 4225 pg/mL (AUC 0.835, sensitivity 98%, specificity 100%, p = 0.014) for the prediction of SO at T3. CONCLUSIONS: MSTN is positively correlated with muscle mass and strength in individuals with cancer or obesity, suggesting its potential use as a biomarker of muscle mass and strength. The ROC curve analysis suggests the potential use of MSTN as a screening tool for CC and SO.


Subject(s)
Biomarkers , Cachexia , Hand Strength , Muscle, Skeletal , Myostatin , Neoplasms , Obesity , Sarcopenia , Humans , Myostatin/blood , Male , Female , Neoplasms/blood , Neoplasms/complications , Neoplasms/physiopathology , Muscle, Skeletal/physiopathology , Middle Aged , Obesity/blood , Obesity/physiopathology , Obesity/complications , Cachexia/blood , Cachexia/etiology , Cachexia/physiopathology , Biomarkers/blood , Sarcopenia/blood , Sarcopenia/etiology , Sarcopenia/physiopathology , Hand Strength/physiology , Body Composition , Aged , Muscle Strength/physiology , Adult , Electric Impedance
2.
Clin Nutr ; 43(6): 1320-1328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669764

ABSTRACT

BACKGROUND & AIMS: GLIM definition of malnutrition is recognised all over the world and, when is referring to cancer, it specifies that weight or muscle loss are associated with an inflammatory status. However, the real-world practice shows that GLIM definition cannot encompass all the wide and heterogenous clinical presentations of cancer patients with malnutrition, which involves many other drivers beyond inflammation. Moreover, placing an excessive emphasis on the inflammation can overshadow, in the clinical practice, the role of the nutritional support in malnourished cancer patients. The aim of this paper is not to criticize the rationale of the GLIM definition of cancer cachexia, but to show the complexity and heterogeneity of malnutrition of cancer patients and reasons why nutritional support should deserve such a better consideration among the oncologists. METHODS: Literature pertinent to pathophysiology of malnutrition of cancer patients is scrutinised and reasons for the frequent underuse of nutritional support are critically analysed. RESULTS: The appraisal of the literature shows that there are various pathophysiological patterns of malnutrition among cancer patients and inflammatory markers are not universally present in weight-losing cancer patients. Inflammation alone does not account for weight loss in all cancer patients and factors other than inflammation can drive hypophagia and weight loss, and hypophagia appears to be a primary catalyst for weight loss. Furthermore, malnutrition may be the consequence of the presence of several Nutrition Impact Symptoms or of the oncologic therapy. The nutritional support may fail to show benefits in malnourished cancer patients because the golden standard to validate a therapy relies on RCT, but it is ethically impossible to have an unfed control group of malnourished patients. Furthermore, nutritional interventions often fell short of the optimal standards, adherence to treatment plans was often poor, nutritional support was mainly reserved for very advanced patients and the primary endpoints of the studies on nutritional support were sometimes unrealistic. CONCLUSION: There is a gap between the suggestion of the guidelines which advocate the use of nutritional support to improve the compliance of patients facing intensive oncologic treatments or to prevent an early demise when patients enter a chronic phase of slow nutritional deterioration, and the poor use of nutrition in the real-world practice. This requires a higher level of awareness of the oncologists concerning the reasons for the lacking evidence of efficacy of the nutritional support and an understanding of its potential contribute to improve the outcome of the patients. Finally, this paper calls for a change of the oncologist's approach to the cancer patient, from only focusing on the cure of the tumour to taking care of the patient as a whole.


Subject(s)
Cachexia , Malnutrition , Neoplasms , Nutritional Status , Nutritional Support , Humans , Malnutrition/therapy , Malnutrition/etiology , Neoplasms/complications , Neoplasms/physiopathology , Cachexia/therapy , Cachexia/etiology , Cachexia/physiopathology , Cachexia/diet therapy , Nutritional Support/methods , Inflammation , Weight Loss
3.
Pharmacol Res ; 203: 107129, 2024 May.
Article in English | MEDLINE | ID: mdl-38461961

ABSTRACT

Cancer-related anorexia-cachexia (CRAC) comprises one of the most common syndromes of advanced cancer patients. The prevalence of CRAC increases from 50% to 80% before death. CRAC is associated not only with impaired quality of life in patients and family members but also with shorter survival. The management of CRAC is a great challenge in clinical practice. There are no definite practice guidelines yet for the prevention and treatment of CRAC. A multimodal strategy is the most effective way to treat anorexia-cachexia. Numerous medications have been suggested and used in clinical trials, while others are still being studied on experimental animals. These medications include branched-chain amino acids, eicosapentaenoic acid, thalidomide, cytokine inhibitors, steroids, antiserotoninergic medications, and appetite stimulants. The benefits of supportive care interventions and the advancement of exciting new pharmacological medicines for anorexia-cachexia are becoming more widely recognized. Health care professionals need to be aware of the psychosocial and biological effects of anorexia-cachexia, even though knowledge of the underlying molecular causes of the disorder has advanced significantly.


Subject(s)
Anorexia , Cachexia , Neoplasms , Humans , Anorexia/therapy , Anorexia/drug therapy , Anorexia/etiology , Anorexia/physiopathology , Neoplasms/complications , Neoplasms/drug therapy , Cachexia/therapy , Cachexia/etiology , Cachexia/physiopathology , Cachexia/drug therapy , Animals
4.
Pancreas ; 53(5): e405-e409, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38517481

ABSTRACT

OBJECTIVES: Pancreatic ductal adenocarcinoma (PDAC) is a common cancer with a poor prognosis and is associated with a high prevalence of cachexia, a metabolic syndrome of muscle wasting due to complex mechanisms. In addition to loss of muscle mass, cancer patients also experience functional deterioration. The aim of this study is to determine whether there is an association between muscle mass and function and clinical outcomes, particularly survival. METHODS: We performed a prospective cohort study including all patients with PDAC at Monash Health from March 2016 to December 2017. We conducted body composition analysis for myopenia and handgrip strength testing. We constructed Kaplan-Meier curves to estimate whether myopenia and low hand grip strength were associated with poorer survival. RESULTS: Myopenia was not associated with a significant difference in PDAC-specific survival (log-rank P = 0.60). However, low handgrip strength was associated with significantly worse PDAC-specific survival compared with other patients (log-rank hazard ratio, 1.88; 95% confidence interval, 1.15-3.09; P = 0.004). CONCLUSIONS: The relationship between survival in PDAC and handgrip strength, but not anatomical muscle mass, suggests that functional testing of strength may be important in prognostication of patients with PDAC, alongside existing tools such as the Eastern Cooperative Oncology Group performance status.


Subject(s)
Carcinoma, Pancreatic Ductal , Hand Strength , Pancreatic Neoplasms , Humans , Hand Strength/physiology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/physiopathology , Male , Female , Aged , Middle Aged , Prospective Studies , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/physiopathology , Prognosis , Body Composition , Kaplan-Meier Estimate , Aged, 80 and over , Cachexia/physiopathology , Cachexia/mortality , Cachexia/diagnosis , Cachexia/etiology
5.
Nat Commun ; 13(1): 149, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013221

ABSTRACT

Cachexia is associated with poor prognosis in chronic heart failure patients, but the underlying mechanisms of cachexia triggered disease progression remain poorly understood. Here, we investigate whether the dysregulation of myokine expression from wasting skeletal muscle exaggerates heart failure. RNA sequencing from wasting skeletal muscles of mice with heart failure reveals a reduced expression of Ostn, which encodes the secreted myokine Musclin, previously implicated in the enhancement of natriuretic peptide signaling. By generating skeletal muscle specific Ostn knock-out and overexpressing mice, we demonstrate that reduced skeletal muscle Musclin levels exaggerate, while its overexpression in muscle attenuates cardiac dysfunction and myocardial fibrosis during pressure overload. Mechanistically, Musclin enhances the abundance of C-type natriuretic peptide (CNP), thereby promoting cardiomyocyte contractility through protein kinase A and inhibiting fibroblast activation through protein kinase G signaling. Because we also find reduced OSTN expression in skeletal muscle of heart failure patients, augmentation of Musclin might serve as therapeutic strategy.


Subject(s)
Cachexia/genetics , Endomyocardial Fibrosis/genetics , Heart Failure/genetics , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Transcription Factors/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/genetics , 2',3'-Cyclic Nucleotide 3'-Phosphodiesterase/metabolism , Aged , Aged, 80 and over , Animals , Cachexia/metabolism , Cachexia/physiopathology , Cachexia/prevention & control , Case-Control Studies , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Disease Models, Animal , Endomyocardial Fibrosis/metabolism , Endomyocardial Fibrosis/physiopathology , Endomyocardial Fibrosis/prevention & control , Female , Gene Expression Regulation , Heart Failure/metabolism , Heart Failure/physiopathology , Heart Failure/prevention & control , Heart Function Tests , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/agonists , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/deficiency , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Muscular Atrophy/prevention & control , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency
6.
Anticancer Res ; 42(1): 397-405, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34969750

ABSTRACT

BACKGROUND/AIM: Cancer cachexia encompasses several deleterious physiological alterations associated with functional impairments, poor quality of life, and increased mortality. The aim of this study was to examine the effects of chronic moderate intensity exercise training on markers of cachexia. MATERIALS AND METHODS: Balb/c mice were randomly assigned to sedentary (SED) or exercise (EX) groups and EX mice were further randomly assigned to one of three exercise modalities (aerobic, resistance, combined). RESULTS: Cachexia was induced in SED animals inoculated with C26 cells, as evidenced by significant changes in numerous markers. All cachexia-related perturbations were significantly attenuated in EX versus SED animals. Systemic inflammation was significantly decreased in all EX groups, as evident by a normalization of spleen mass and plasma IL-6. CONCLUSION: Multiple moderate intensity exercise modalities can provide significant benefits in cachectic mice, and this may be due, at least in part, to decreased systemic inflammation.


Subject(s)
Cachexia/therapy , Exercise/physiology , Neoplasms/therapy , Physical Conditioning, Animal , Animals , Cachexia/etiology , Cachexia/physiopathology , Disease Models, Animal , Humans , Mice , Muscle, Skeletal/physiology , Neoplasms/complications , Neoplasms/physiopathology , Physical Therapy Modalities , Quality of Life , Resistance Training , Sedentary Behavior
7.
Sci Rep ; 11(1): 23627, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34880268

ABSTRACT

Cachexia is a muscle wasting syndrome occurring in many advanced cancer patients. Cachexia significantly increases cancer morbidity and mortality. Cardiac atrophy and contractility deficits have been observed in patients and in animal models with cancer cachexia, which may contribute to cachexia pathophysiology. However, underlying contributors to decreased in vivo cardiac contractility are not well understood. In this study, we sought to distinguish heart-intrinsic changes from systemic factors contributing to cachexia-associated cardiac dysfunction. We hypothesized that isolated heart and cardiac myocyte functional deficits underlie in vivo contractile dysfunction. To test this hypothesis, isolated heart and cardiac myocyte function was measured in the colon-26 adenocarcinoma murine model of cachexia. Ex vivo perfused hearts from cachectic animals exhibited marked contraction and relaxation deficits during basal and pacing conditions. Isolated myocytes displayed significantly decreased peak contraction and relaxation rates, which was accompanied by decreased peak calcium and decay rates. This study uncovers significant organ and cellular-level functional deficits in cachectic hearts outside of the catabolic in vivo environment, which is explained in part by impaired calcium cycling. These data provide insight into physiological mechanisms of cardiomyopathy in cachexia, which is critical for the ultimate development of effective treatments for patients.


Subject(s)
Cachexia/physiopathology , Calcium/metabolism , Heart Failure/etiology , Myocardial Contraction , Myocytes, Cardiac/pathology , Neoplasms, Experimental/physiopathology , Animals , Body Weight , Cachexia/complications , Cell Line, Tumor , Humans , Male , Mice , Muscular Atrophy/metabolism , Myocytes, Cardiac/metabolism , Neoplasms, Experimental/complications , Organ Size
8.
Int J Mol Sci ; 22(24)2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34948370

ABSTRACT

Muscle deconditioning impairs both locomotor function and metabolic health, and is associated with reduced quality life and increased mortality rates. Despite an appreciation of the existence of phenomena such as muscle anabolic resistance, mitophagy, and insulin resistance with age and disease in humans, little is known about the mechanisms responsible for these negative traits. With the complexities surrounding these unknowns and the lack of progress to date in development of effective interventions, there is a need for alternative approaches. Metabolomics is the study of the full array of metabolites within cells or tissues, which collectively constitute the metabolome. As metabolomics allows for the assessment of the cellular metabolic state in response to physiological stimuli, any chronic change in the metabolome is likely to reflect adaptation in the physiological phenotype of an organism. This, therefore, provides a holistic and unbiased approach that could be applied to potentially uncover important novel facets in the pathophysiology of muscle decline in ageing and disease, as well as identifying prognostic markers of those at risk of decline. This review will aim to highlight the current knowledge and potential impact of metabolomics in the study of muscle mass loss and deconditioning in humans and will highlight key areas for future research.


Subject(s)
Aging , Metabolome , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Amino Acids/metabolism , Animals , Cachexia/metabolism , Cachexia/physiopathology , Energy Metabolism , Humans , Lipid Metabolism , Metabolomics , Muscle, Skeletal/physiopathology , Sarcopenia/physiopathology
9.
Dev Cell ; 56(19): 2741-2751.e7, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34610327

ABSTRACT

Cancer cachexia is associated with many types of tumors and is characterized by a combination of anorexia, loss of body weight, catabolic alterations, and systemic inflammation. We developed a tumor model in Drosophila larvae that causies cachexia-like syndrome, and we found that cachectic larvae show reduced levels of the circulating steroid ecdysone (Ec). Artificially importing Ec in the tumor through the use of the EcI/Oatp74D importer aggravated cachexia, whereas feeding animals with Ec rescued cachectic defects. This suggests that a steroid sink induced by the tumor promotes catabolic alterations in healthy tissues. We found that Oatp33Eb, a member of the Oatp transporter family, is specifically induced in tumors promoting cachexia. The overexpression of Oatp33Eb in noncachectic tumors induced cachexia, whereas its inhibition in cachectic tumors restored circulating Ec and reversed cachectic alterations. Oatp transporters are induced in several types of hormone-dependent tumors, and this result suggests that a similar sink effect could modify hormonal balance in cachectic cancer patients.


Subject(s)
Cachexia/metabolism , Ecdysone/metabolism , Organic Anion Transporters/metabolism , Animals , Body Weight , Cachexia/physiopathology , Drosophila Proteins , Drosophila melanogaster , Larva/metabolism , Neoplasms , Organic Anion Transporters/physiology , Steroids/metabolism
10.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502376

ABSTRACT

Inflammation induces a wide response of the neuroendocrine system, which leads to modifications in all the endocrine axes. The hypothalamic-growth hormone (GH)-insulin-like growth factor-1 (IGF-1) axis is deeply affected by inflammation, its response being characterized by GH resistance and a decrease in circulating levels of IGF-1. The endocrine and metabolic responses to inflammation allow the organism to survive. However, in chronic inflammatory conditions, the inhibition of the hypothalamic-GH-IGF-1 axis contributes to the catabolic process, with skeletal muscle atrophy and cachexia. Here, we review the changes in pituitary GH secretion, IGF-1, and IGF-1 binding protein-3 (IGFBP-3), as well as the mechanism that mediated those responses. The contribution of GH and IGF-1 to muscle wasting during inflammation has also been analyzed.


Subject(s)
Cachexia/metabolism , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor I/metabolism , Cachexia/physiopathology , Growth Hormone/metabolism , Human Growth Hormone/metabolism , Humans , Hypothalamus/metabolism , Inflammation/physiopathology , Insulin/metabolism , Insulin-Like Growth Factor Binding Protein 3/physiology , Insulin-Like Growth Factor I/physiology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology
11.
Molecules ; 26(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34443492

ABSTRACT

Muscle wasting and cachexia are prominent comorbidities in cancer. Treatment with polyphenolic compounds may partly revert muscle wasting. We hypothesized that treatment with curcumin or resveratrol in cancer cachectic mice may improve muscle phenotype and total body weight through attenuation of several proteolytic and signaling mechanisms in limb muscles. In gastrocnemius and soleus muscles of cancer cachectic mice (LP07 adenocarcinoma cells, N = 10/group): (1) LC-induced cachexia, (2) LC-cachexia+curcumin, and (3) LC-cachexia + resveratrol, muscle structure and damage (including blood troponin I), sirtuin-1, proteolytic markers, and signaling pathways (NF-κB and FoxO3) were explored (immunohistochemistry and immunoblotting). Compared to nontreated cachectic mice, in LC-cachexia + curcumin and LC-cachexia + resveratrol groups, body and muscle weights (gastrocnemius), limb muscle strength, muscle damage, and myofiber cross-sectional area improved, and in both muscles, sirtuin-1 increased, while proteolysis (troponin I), proteolytic markers, and signaling pathways were attenuated. Curcumin and resveratrol elicited beneficial effects on fast- and slow-twitch limb muscle phenotypes in cachectic mice through sirtuin-1 activation, attenuation of atrophy signaling pathways, and proteolysis in cancer cachectic mice. These findings have future therapeutic implications as these natural compounds, separately or in combination, may be used in clinical settings of muscle mass loss and dysfunction including cancer cachexia.


Subject(s)
Cachexia/etiology , Cachexia/physiopathology , Curcumin/pharmacology , Muscles/pathology , Muscles/physiopathology , Neoplasms/complications , Proteolysis , Resveratrol/pharmacology , Animals , Biomarkers/metabolism , Cell Line , Female , Mice, Inbred BALB C , Muscle Proteins/metabolism , Muscles/drug effects , Muscular Atrophy/metabolism , Phenotype , Proteolysis/drug effects , Signal Transduction , Sirtuin 1/metabolism
12.
Int J Mol Sci ; 22(16)2021 Aug 06.
Article in English | MEDLINE | ID: mdl-34445197

ABSTRACT

The term "cachexia" is derived from the Greek words kakos (bad) and hexis (habit). Cachexia is a malnutrition associated with chronic diseases such as cancer, chronic heart failure, chronic renal failure, and autoimmune diseases, and is characterized by decreased skeletal muscle mass. Cancer cachexia is quite common in patients with advanced cancer. Weight loss is also a characteristic symptom of cancer cachexia, along with decreased skeletal muscle mass. As nutritional supplementation alone cannot improve cachexia, cytokines and tumor-derived substances have been attracting attention as its relevant factors. Cancer cachexia can be also associated with reduced chemotherapeutic effects, increased side effects and treatment interruptions, and even poorer survival. In 2011, a consensus definition of cachexia has been proposed, and the number of relevant research reports has increased significantly. However, the pathogenesis of cachexia is not fully understood, and there are currently few regulatory-approved standard treatments for cachexia. The main reason for this is that multiple etiologies are involved in the development of cachexia. In this review, we will outline the current status of cachexia, the mechanisms of which have been elucidated in recent years, especially from the perspective of advanced cancer.


Subject(s)
Cachexia/etiology , Neoplasms/complications , Anilides/therapeutic use , Animals , Cachexia/diagnosis , Cachexia/physiopathology , Cachexia/therapy , Dietary Supplements , Disease Management , Humans , Hydrazines/therapeutic use , Neoplasms/physiopathology , Oligopeptides/therapeutic use
13.
Pancreas ; 50(5): 657-666, 2021.
Article in English | MEDLINE | ID: mdl-34106574

ABSTRACT

ABSTRACT: Diet and exercise interventions may help reverse malnutrition and muscle wasting common in pancreatic cancer. We performed a scoping review to identify the knowledge gaps surrounding diet and exercise interventions. We searched PubMed, Scopus, Cumulative Index to Nursing and Allied Health Literature, Embase, ProQuest Theses and Dissertations, and Google Scholar using the umbrella terms of "pancreatic cancer," "diet/nutrition," and "exercise." Included were articles reporting on ambulatory adults with diagnosed pancreatic cancer. Excluded were studies examining prevention and/or risk, animal, or cell lines. Of the 15,708 articles identified, only 62 met the final inclusion criteria. Almost half of the articles were randomized controlled studies (n = 27). Most studies were from the United States (n = 20). The majority examined dietary interventions (n = 41), with 20 assessing the use of omega-3 fatty acids. Exercise interventions were reported in 13 studies, with 8 examining a diet and exercise intervention. Most studies were small and varied greatly in terms of study design, intervention, and outcomes. We identified 7 research gaps that should be addressed in future studies. This scoping review highlights the limited research examining the effect of diet and exercise interventions in ambulatory patients with pancreatic cancer.


Subject(s)
Cachexia/diet therapy , Exercise Therapy , Malnutrition/diet therapy , Muscular Atrophy/diet therapy , Nutrition Therapy , Pancreatic Neoplasms/diet therapy , Body Composition , Cachexia/epidemiology , Cachexia/physiopathology , Diet, Healthy , Dietary Supplements/adverse effects , Humans , Malnutrition/epidemiology , Malnutrition/physiopathology , Muscle Strength , Muscle, Skeletal/physiopathology , Muscular Atrophy/epidemiology , Muscular Atrophy/physiopathology , Nutritional Status , Nutritive Value , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/physiopathology , Treatment Outcome
14.
J Appl Toxicol ; 41(12): 1952-1965, 2021 12.
Article in English | MEDLINE | ID: mdl-33977518

ABSTRACT

Allograft and xenograft transplantation into a mouse host is frequently utilized to study cancer biology, tumor behavior, and response to treatment. Preclinical studies employing these models often focus solely upon the intra-tumoral effects of a given treatment, without consideration of systemic toxicity or tumor-host interaction, nor whether this latter relationship could modulate the toxicologic response to therapy. Here it is demonstrated that the implantation and growth of a range of human- and mouse-derived cell lines leads to structural vascular and, potentially, functional changes within peripheral endocrine tissues, a process that could conceivably ameliorate the severity of anti-angiogenic-induced fenestrated vessel attenuation. Observations suggest a multifactorial process, which may involve host- and tumor-derived cytokines/growth factors, and the liberation of myeloid-derived suppressor cells. Further investigation revealed a structurally comparable response to the administration of exogenous estrogen. These findings, in addition to providing insight into the development of clinical anti-angiogenic "adaptation," may be of significance within the "cancer-cachexia" and cancer-related anemia syndromes in man.


Subject(s)
Anemia/physiopathology , Cachexia/physiopathology , Cytokines/metabolism , Endocrine System/physiopathology , Animals , Cell Line, Tumor , Mice , Neoplasms/physiopathology
15.
Int J Mol Sci ; 22(9)2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33925872

ABSTRACT

Cancer cachexia is a common condition in many cancer patients, particularly those with advanced disease. Cancer cachexia patients are generally less tolerant to chemotherapies and radiotherapies, largely limiting their treatment options. While the search for treatments of this condition are ongoing, standards for the efficacy of treatments have yet to be developed. Current diagnostic criteria for cancer cachexia are primarily based on loss of body mass and muscle function. However, these criteria are rather limiting, and in time, when weight loss is noticeable, it may be too late for treatment. Consequently, biomarkers for cancer cachexia would be valuable adjuncts to current diagnostic criteria, and for assessing potential treatments. Using high throughput methods such as "omics approaches", a plethora of potential biomarkers have been identified. This article reviews and summarizes current studies of biomarkers for cancer cachexia.


Subject(s)
Biomarkers, Tumor , Cachexia , Neoplasms/complications , Biomarkers/blood , Biomarkers/metabolism , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Cachexia/diagnosis , Cachexia/physiopathology , Humans , Muscle, Skeletal/physiopathology , Risk Factors , Weight Loss
17.
J Cancer Res Clin Oncol ; 147(5): 1443-1450, 2021 May.
Article in English | MEDLINE | ID: mdl-33718995

ABSTRACT

PURPOSE: Cancer-associated cachexia is a common condition in patients with advanced cancer, and is associated with extreme and involuntary weight loss and irreversible muscle wasting. Despite its high morbidity and mortality, there is no known treatment to reverse its effects. Thus, there is increasing interest in whether diet and exercise can assist in the minimization of cancer-associated cachexia. METHODS: We reviewed the literature on the impact of dietary patterns, dietary components, and exercise on the progress and severity of cancer cachexia. RESULTS: Although most studies have produced inconclusive or controversial findings, some promising studies using animal models and early human clinical trials suggest that dietary and physical therapy interventions may alleviate cancer-associated cachexia. Moreover, many studies suggest that controlling diet and exercise nevertheless improved the quality of life (QoL) for cancer patients with cachexia. CONCLUSION: Ongoing studies will continue to examine whether different forms of multimodal therapy-combinations of cancer treatment, dietary regimens, anti-inflammatory therapy, and physical therapy-are effective methods to improve outcomes in advanced cancer patients with cachexia. Moreover, future studies should examine the effects of such interventions on long-term QoL and establish nutritional guidelines for the management of cancer-associated cachexia.


Subject(s)
Cachexia/genetics , Neoplasms/complications , Neoplasms/physiopathology , Animals , Cachexia/physiopathology , Diet , Humans , Physical Therapy Modalities , Quality of Life
18.
Cells ; 10(3)2021 02 28.
Article in English | MEDLINE | ID: mdl-33671024

ABSTRACT

Muscle wasting, i.e., cachexia, frequently occurs in cancer and associates with poor prognosis and increased morbidity and mortality. Anticancer treatments have also been shown to contribute to sustainment or exacerbation of cachexia, thus affecting quality of life and overall survival in cancer patients. Pre-clinical studies have shown that blocking activin receptor type 2 (ACVR2) or its ligands and their downstream signaling can preserve muscle mass in rodents bearing experimental cancers, as well as in chemotherapy-treated animals. In tumor-bearing mice, the prevention of skeletal and respiratory muscle wasting was also associated with improved survival. However, the definitive proof that improved survival directly results from muscle preservation following blockade of ACVR2 signaling is still lacking, especially considering that concurrent beneficial effects in organs other than skeletal muscle have also been described in the presence of cancer or following chemotherapy treatments paired with counteraction of ACVR2 signaling. Hence, here, we aim to provide an up-to-date literature review on the multifaceted anti-cachectic effects of ACVR2 blockade in preclinical models of cancer, as well as in combination with anticancer treatments.


Subject(s)
Activin Receptors, Type II/metabolism , Cachexia/etiology , Neoplasms/complications , Cachexia/physiopathology , Humans , Neoplasms/mortality , Signal Transduction , Survival Analysis
19.
J Biomed Sci ; 28(1): 15, 2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33658022

ABSTRACT

Although rheumatoid arthritis affects 1% of the global population, the role of rheumatoid cachexia, which occurs in up to a third of patients, is relatively neglected as research focus, despite its significant contribution to decreased quality of life in patients. A better understanding of the cellular and molecular processes involved in rheumatoid cachexia, as well as its potential treatment, is dependent on elucidation of the intricate interactions of the cells involved, such as myoblasts, fibroblasts and macrophages. Persistent RA-associated inflammation results in a relative depletion of the capacity for regeneration and repair in the satellite cell niche. The repair that does proceed is suboptimal due to dysregulated communication from the other cellular role players in this multi-cellular environment. This includes the incomplete switch in macrophage phenotype resulting in a lingering pro-inflammatory state within the tissues, as well as fibroblast-associated dysregulation of the dynamic control of the extracellular matrix. Additional to this endogenous dysregulation, some treatment strategies for RA may exacerbate muscle wasting and no multi-cell investigation has been done in this context. This review summarizes the most recent literature characterising clinical RA cachexia and links these features to the roles of and complex communication between multiple cellular contributors in the muscle niche, highlighting the importance of a targeted approach to therapeutic intervention.


Subject(s)
Arthritis, Rheumatoid/complications , Cachexia/physiopathology , Fibroblasts/metabolism , Macrophages/metabolism , Muscle, Skeletal/pathology , Myoblasts/metabolism , Animals , Cachexia/etiology , Humans , Mice , Rats
20.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540821

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related death, and the prevalence of CRC in young adults is on the rise, making this a largescale clinical concern. Advanced CRC patients often present with liver metastases (LM) and an increased incidence of cachexia, i.e., musculoskeletal wasting. Despite its high incidence in CRC patients, cachexia remains an unresolved issue, and animal models for the study of CRC cachexia, in particular, metastatic CRC cachexia, remain limited; therefore, we aimed to establish a new model of metastatic CRC cachexia. C57BL/6 male mice (8 weeks old) were subcutaneously (MC38) or intrasplenically injected (mMC38) with MC38 murine CRC cells to disseminate LM, while experimental controls received saline (n = 5-8/group). The growth of subcutaneous MC38 tumors was accompanied by a reduction in skeletal muscle mass (-16%; quadriceps muscle), plantarflexion force (-22%) and extensor digitorum longus (EDL) contractility (-20%) compared to experimental controls. Meanwhile, the formation of MC38 LM (mMC38) led to heighted reductions in skeletal muscle mass (-30%; quadriceps), plantarflexion force (-28%) and EDL contractility (-35%) compared to sham-operated controls, suggesting exacerbated cachexia associated with LM. Moreover, both MC38 and mMC38 tumor hosts demonstrated a marked loss of bone indicated by reductions in trabecular (Tb.BV/TV: -49% in MC38, and -46% in mMC38) and cortical (C.BV/TV: -12% in MC38, and -8% in mMC38) bone. Cell culture experiments revealed that MC38 tumor-derived factors directly promote myotube wasting (-18%) and STAT3 phosphorylation (+5-fold), while the pharmacologic blockade of STAT3 signaling was sufficient to preserve myotube atrophy in the presence of MC38 cells (+21%). Overall, these results reinforce the notion that the formation of LM heightens cachexia in an experimental model of CRC.


Subject(s)
Adenocarcinoma/secondary , Cachexia/etiology , Colorectal Neoplasms/complications , Liver Neoplasms/secondary , Adenocarcinoma/complications , Adenocarcinoma/pathology , Animals , Cachexia/pathology , Cachexia/physiopathology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Progression , Femur/pathology , Liver Neoplasms/complications , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Muscle Contraction , Muscle Fibers, Skeletal/ultrastructure , Muscle Weakness/etiology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy/etiology , STAT3 Transcription Factor , Subcutaneous Tissue , X-Ray Microtomography
SELECTION OF CITATIONS
SEARCH DETAIL
...