1
|
Barone M, Baccaro P, Molfino A. An Overview of Sarcopenia: Focusing on Nutritional Treatment Approaches. Nutrients 2025; 17:1237. [PMID: 40218995 PMCID: PMC11990658 DOI: 10.3390/nu17071237] [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: 03/09/2025] [Revised: 03/28/2025] [Accepted: 03/30/2025] [Indexed: 04/14/2025] Open
Abstract
Sarcopenia is a syndrome characterized by the progressive and generalized loss of skeletal muscle mass and strength. This condition is associated with physical disability, decreased quality of life, and increased mortality. Therefore, reducing the prevalence of sarcopenia could significantly lower healthcare costs. Sarcopenia can be classified into primary and secondary sarcopenia. The former is related to aging and begins after the fourth decade of life; after that, there is a muscle loss of around 8% per decade until age 70 years, which subsequently increases to 15% per decade. On the other hand, secondary sarcopenia can affect all individuals and may result from various factors including physical inactivity, malnutrition, endocrine disorders, neurodegenerative diseases, inflammation, and cachexia. Understanding the multiple mechanisms involved in the onset and progression of sarcopenia allows for us to develop strategies that can prevent, treat, or at least mitigate muscle loss caused by increased protein breakdown. One potential treatment of sarcopenia is based on nutritional interventions, including adequate caloric and protein intake and specific nutrients that support muscle health. Such nutrients include natural food rich in whey protein and omega-3 fatty acids as well as nutritional supplements like branched-chain amino acids, β-hydroxy-β-methylbutyrate, and vitamin D along with food for special medical purposes. It is important to emphasize that physical exercises, especially resistance training, not only promote muscle protein synthesis on their own but also work synergistically with nutritional strategies to enhance their effectiveness.
Collapse
Affiliation(s)
- Michele Barone
- Gastroenterology Unit, Department of Precision and Regenerative Medicine, University of Bari, Policlinic University Hospital, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Palmina Baccaro
- Gastroenterology Unit, Department of Precision and Regenerative Medicine, University of Bari, Policlinic University Hospital, Piazza G. Cesare 11, 70124 Bari, Italy;
| | - Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| |
Collapse
|
2
|
Kawabata R, Nishikawa K, Kawase T, Kawada J, Kimura Y, Kashima Y, Ueda S, Takeno A, Shimomura K, Imamura H. Multicenter phase II study on the efficacy of an oral nutritional supplement containing eicosapentaenoic acid in advanced gastric cancer patients with cachexia. Gastric Cancer 2025:10.1007/s10120-025-01605-x. [PMID: 40106056 DOI: 10.1007/s10120-025-01605-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Cachexia is a common complication in advanced gastric cancer (AGC). Eicosapentaenoic acid (EPA) may ameliorate cachexia. This single-arm, phase II study assessed the potential benefit of an oral nutritional supplement containing EPA (ONS-EPA) for cachexia in AGC patients. METHODS Chemotherapy-naive AGC patients with cachexia, defined by serum albumin < 3.5 g/dl and C-reactive protein ≥ 0.5 mg/dl, were included. Patients received an EPA-enriched supplement (Prosure®, 1.056 g EPA/pack) twice daily during first-line chemotherapy. The primary endpoint was time to treatment failure (TTF) in patients adhering to ≥ 25% of the planned ONS-EPA dose in the first two weeks (per-protocol set, PPS). Secondary analyses evaluated adherence impact on treatment outcomes. RESULTS Of 72 enrolled patients, 65 were evaluated. Median adherence was 42.8%. Median TTF in the PPS group was 4.8 months (95% CI 3.6-5.5), below the pre-set 4-month threshold. The PPS group had a higher proportion of patients who improved their nutritional and inflammatory status during treatment, along with better TTF and overall survival (OS) compared to those with poor adherence. Adjusted median TTF was 4.6 vs. 2.5 months (hazard ratio: 0.56; 95% CI 0.28-1.12, p = 0.105). CONCLUSIONS Although the primary endpoint was not achieved, the study suggests that ONS-EPA may benefit AGC patients with cachexia.
Collapse
Affiliation(s)
- Ryohei Kawabata
- Department of Surgery, Sakai City Medical Center, 1-1-1, Ebaraji-cho, Nishi-ku, Sakai, Osaka, 5938304, Japan.
- Department of Surgery, Osaka Rosai Hospital, Sakai, Japan.
| | - Kazuhiro Nishikawa
- Department of Surgery, Osaka General Medical Center, Osaka, Japan
- Cancer Treatment Center, Osaka International Medical and Science Center, Osaka Keisatsu Hospital, Osaka, Japan
| | - Tomono Kawase
- Department of Surgery, Sakai City Medical Center, 1-1-1, Ebaraji-cho, Nishi-ku, Sakai, Osaka, 5938304, Japan
- Department of Surgery, Toyonaka Municipal Hospital, Toyonaka, Japan
| | - Junji Kawada
- Department of Surgery, Osaka General Medical Center, Osaka, Japan
| | - Yutaka Kimura
- Department of Surgery, Sakai City Medical Center, 1-1-1, Ebaraji-cho, Nishi-ku, Sakai, Osaka, 5938304, Japan
| | | | - Shugo Ueda
- Department of Gastroenterological Surgery, Kitano Hospital, Osaka, Japan
| | - Atsushi Takeno
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Japan
| | | | - Hiroshi Imamura
- Department of Surgery, Sakai City Medical Center, 1-1-1, Ebaraji-cho, Nishi-ku, Sakai, Osaka, 5938304, Japan
- Cancer Treatment Center, Osaka International Medical and Science Center, Osaka Keisatsu Hospital, Osaka, Japan
| |
Collapse
|
3
|
Vitale E, Maistrello L, Rizzo A, Brunetti O, Massafra R, Mollica V, Massari F, Santoni M. Nutritional conditions and PFS and OS in cancer immunotherapy: the MOUSEION-010 meta-analysis. Immunotherapy 2025; 17:269-281. [PMID: 40134096 PMCID: PMC12013447 DOI: 10.1080/1750743x.2025.2483656] [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: 11/07/2024] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The MOUSEION-010 Meta-Analysis assessed the association between nutritional status and clinical outcomes such as Progression Free Survival (PFS) and Overall Survival (OS) among cancer patients treated with immune checkpoint inhibitors (ICIs). METHODS Nutritional status was assessed based on the Prognostic Nutrition Index (PNI), Geriatric Nutritional Risk Index (GNRI) and Controlling Nutritional Status (CONUT) indexes. Databases consulted were: Embase, PubMed, Scopus and Web of Science. RESULTS PNI and GNRI indexes did not show a significant association with both PFS and OS, while CONUT index displayed a significant difference in PFS between the two groups, in favor of the control group (Z = 4.04; p < 0.01) also without any publication bias (β= -1.27; 95% CI = [-2.13; -0.42]; p = 0.10]). The same trend was recorded in OS, too (Z = 4.24; p < 0.01). However, publication bias was present (β = 1.89; 95% CI = [1.26; 2.54]; p = 0.028]) and the numerosity of the studies did not reveal the sufficient statistical power to obtain reliable results. CONCLUSION Malnutrition could negatively impact cancer patients, especially in advanced phases. Our findings could be associated with the reduction of physical ability and daily activity performance, lower compliance with treatment protocols, and shorter survival outcomes.
Collapse
Affiliation(s)
- Elsa Vitale
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Oronzo Brunetti
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Raffaella Massafra
- Scientific Directorate, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | | |
Collapse
|
4
|
You Y, Wang Y, Zhang G, Li Y. The Molecular Mechanisms and Treatment of Cancer-Related Cachexia. J Nutr Sci Vitaminol (Tokyo) 2025; 71:1-15. [PMID: 40024744 DOI: 10.3177/jnsv.71.1] [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: 03/04/2025]
Abstract
Cancer cachexia is a multifactorial syndrome characterized by persistent skeletal muscle loss, with or without fat loss, which cannot be completely reversed by traditional nutritional support and leads to impaired organ function. Cachexia seriously reduces the quality of life of (QOL) patients, affects the therapeutic effect against cancers, increases the incidence of complications, and is an important cause of death for patients with advanced cancers. To date, no effective medical intervention has completely reversed cachexia, and no medication has been agreed upon. Here, we describe recent advances in the diagnosis, molecular mechanism and treatment of cancer-related cachexia.
Collapse
Affiliation(s)
- Yongfei You
- Department of Medical Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University
- Department of Medical Oncology, The Third Affiliated Hospital, Jiangxi Medical College, Nanchang University, The First Hospital of Nanchang
| | - Yong Wang
- Department of Medical Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University
| | - Guohua Zhang
- Nanchang Key Laboratory of Tumor Gene Diagnosis and Innovative Treatment Research, Gaoxin Branch of the First Affiliated Hospital, Jiangxi Medical College, Nanchang University
| | - Yong Li
- Department of Medical Oncology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University
| |
Collapse
|
5
|
Nakamura K, Takahashi Y, Yanai Y, Miyagi T, Nakano H, Koyama Y, Hashimoto H. Fat-free oral nutritional supplements for patients after acute illness: a prospective observational study. J Clin Biochem Nutr 2025; 76:85-89. [PMID: 39896161 PMCID: PMC11782773 DOI: 10.3164/jcbn.24-162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 09/27/2024] [Indexed: 02/04/2025] Open
Abstract
There is a lack of evidence for compliance with and the acceptability of oral nutritional supplements (ONS) by post-acute care patients. Therefore, the present study examined compliance with fat-free ONS, which are easy to drink. Patients who started oral intake in the general ward after being transferred from the Emergency Department were offered three ONS including fat-free ONS: Isocal Clear, Maybalance Mini, and Medimil, three times a day for three days. On days 1 and 3, patients evaluated each ONS using a questionnaire. Thirty-five eligible patients participated in the present study, which began a median of 10 days after their admission. Median taste ratings for Isocal Clear, Maybalance, and Medimil on day 1 were 8, 7, and 3, respectively, while median ease-to-drink ratings were 8, 7, and 5, respectively. In contrast, median taste ratings on day 3 were 5, 0, and 0, respectively, while median ease-to-drink ratings were 7, 1, and 0, respectively. Intakes of the prescribed diet during the three days had a median value as low as 30-50%. In conclusion, good compliance with fat-free ONS by post-acute care patients may be achieved.
Collapse
Affiliation(s)
- Kensuke Nakamura
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, 2-1-1, Jonan-cho, Hitachi, Ibaraki 317-0077, Japan
| | - Yuji Takahashi
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Yusuke Yanai
- Nutrition Department, Hitachi General Hospital, 2-1-1, Jonan-cho, Hitachi, Ibaraki 317-0077, Japan
| | - Tomoka Miyagi
- Department of Emergency and Critical Care Medicine, Hitachi General Hospital, 2-1-1, Jonan-cho, Hitachi, Ibaraki 317-0077, Japan
| | - Hidehiko Nakano
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Yasuaki Koyama
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| | - Hideki Hashimoto
- Department of Critical Care Medicine, Yokohama City University Hospital, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa 236-0004, Japan
| |
Collapse
|
6
|
Conforto R, Rizzo V, Russo R, Mazza E, Maurotti S, Pujia C, Succurro E, Arturi F, Ferro Y, Sciacqua A, Pujia A, Montalcini T. Advances in body composition and gender differences in susceptibility to frailty syndrome: Role of osteosarcopenic obesity. Metabolism 2024; 161:156052. [PMID: 39490438 DOI: 10.1016/j.metabol.2024.156052] [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: 08/05/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
There is general consensus that an improper diet negatively impacts health and that nutrition is a primary tool for the prevention of non-communicable diseases. Unfortunately, the importance of studying body composition, which can reveal early predictors of gender-related diseases, is still not well understood in this context. Currently, individuals are still classified as obese based solely on their body mass index, without considering the amount of fat, its distribution, and the quantity of muscle and bone mass. In this regard, the body composition phenotype defined as "osteosarcopenic obesity" affects approximately 6-41 % of postmenopausal women, with prevalence increasing with age due to the hormonal and metabolic changes that occur during this period. This particular phenotype arises from the strong relationship between visceral fat, muscle, bone, and gut microbiota and predispose postmenopausal women to frailty. Frailty is a complex clinical phenomenon with significant care and economic implications for our society. Recent studies suggest that women have a higher prevalence of frailty syndrome and its individual components, such as osteoporosis, fractures and sarcopenia, compared to men. Here, we provide a comprehensive overview of recent advances regarding the impact of gender on body composition and frailty. Furthermore, we reflect on the crucial importance of personalized nutritional interventions, with a focus on reducing visceral fat, increasing protein intake and optimizing vitamin D levels. A review of the scientific literature on this topic highlights the importance of studying body composition for a personalized and gender-specific approach to nutrition and dietetics, in order to identify frailty syndrome early and establish personalized treatments. This new method of researching disease predictors could likely help clarify the controversial results of studies on vitamin D, calcium and proteins, translate into practical wellness promotion across diverse elderly populations.
Collapse
Affiliation(s)
- Rosy Conforto
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Valeria Rizzo
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Raffaella Russo
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Elisa Mazza
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Samantha Maurotti
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy.
| | - Carmelo Pujia
- O.U. Clinical Nutrition, Renato Dulbecco Hospital, 88100 Catanzaro, Italy
| | - Elena Succurro
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Franco Arturi
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Yvelise Ferro
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy; Research Center for the Prevention and Treatment of Metabolic Diseases, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, University "Magna Græcia" of Catanzaro, 88100 Catanzaro, Italy; Research Center for the Prevention and Treatment of Metabolic Diseases, University "Magna Græcia", 88100 Catanzaro, Italy
| |
Collapse
|
7
|
Purcell SA, Craven SA, Limon-Miro AT, Elliott SA, Melanson EL, Tandon P, Prado CM. Total energy expenditure measured using doubly labeled water in adults with major chronic diseases: a systematic review. Am J Clin Nutr 2024; 120:1071-1084. [PMID: 39209153 PMCID: PMC11600028 DOI: 10.1016/j.ajcnut.2024.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Energy requirement assessment is a cornerstone for nutrition practice. The extent to which total energy expenditure (TEE; indicator of energy requirements) has been measured in adults with chronic diseases has not been explored. OBJECTIVES This systematic review aimed to characterize evidence on TEE among individuals with chronic diseases and describe TEE across chronic diseases and in comparison to controls without a chronic disease. METHODS A literature search using terms related to doubly labeled water and TEE was conducted in PubMed, MEDLINE, Web of Science, and Embase. Eligible articles included those that measured TEE using doubly labeled water in adults with a major chronic disease. Methodological quality was determined using the Academy of Nutrition and Dietetics quality criteria checklist. Sample size-weighted TEE was calculated in each chronic disease subgroup. RESULTS Fifty studies were included, of which 15 had a control group. Median sample size was 20 participants, and approximately half of studies were published over 10 y ago. Thirty-five (70%) studies reported resting energy expenditure, and approximately half (k = 26) reported physical activity level. Methodological quality was neutral (k = 25) or positive (k = 23) for most studies. TEE among individual studies ranged from 934 to 3274 kcal/d. Mean weighted TEE was lowest among gastrointestinal (1786 kcal/d) and neurologic (2104 kcal/d) subgroups and highest among cancer (2903 kcal/d), endocrine (2661 kcal/d), and autoimmune (2625 kcal/d) subgroups. Excluding 1 article in cancer survivors resulted in a low TEE in the cancer subgroup (2112 kcal/d). Most studies with a control group reported no differences in TEE between controls and patients; however, only 1 study was powered for between-group comparisons. CONCLUSIONS Energy requirements vary across chronic diseases, although there is insufficient evidence to suggest that TEE in patients with chronic disease is different than that among controls. Further research is needed to inform energy requirement recommendations that consider chronic disease. This review was registered at PROSPERO as CRD42022336500 (https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=336500).
Collapse
Affiliation(s)
- Sarah A Purcell
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Centre for Chronic Disease Prevention and Management, Faculty of Medicine, University of British Columbia, Kelowna, British Columbia, Canada; School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada.
| | - Sarah A Craven
- School of Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, British Columbia, Canada
| | - Ana Teresa Limon-Miro
- Division of Gastroenterology (Liver Unit), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Human Nutrition Research Unit, Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Sarah A Elliott
- Alberta Research Centre for Health Evidence, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada; Cochrane Child Health, Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Edward L Melanson
- Division of Endocrinology Metabolism and Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States; Division of Geriatric Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Puneeta Tandon
- Division of Gastroenterology (Liver Unit), Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Carla M Prado
- Human Nutrition Research Unit, Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
8
|
Deng J, Wu S, Huang Y, Deng Y, Yu K. Esophageal cancer risk is influenced by genetically determined blood metabolites. Medicine (Baltimore) 2024; 103:e40122. [PMID: 39470544 PMCID: PMC11521038 DOI: 10.1097/md.0000000000040122] [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: 05/24/2024] [Revised: 07/13/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
It remains unclear what causes esophageal cancer (EC), but blood metabolites have been connected to it. Our study performed a Mendelian randomization (MR) analysis to assess the causality from genetically proxied 1400 blood metabolites to EC level. A two-sample MR analysis was employed to evaluate the causal relationship between 1400 blood metabolites and EC. Initially, the EC genome-wide association study (GWAS) data (from Jiang L et al) were examined, leading to the identification of certain metabolites. Subsequently, another set of EC GWAS data from FINNGEN was utilized to validate the findings. Causality was primarily determined through inverse variance weighting, with additional support from the MR-Egger, weighted median, and MR-PRESSO models. Heterogeneity was assessed using the MR Cochran Q test. The MR-Egger intercept and MR-PRESSO global methods were employed to detect multicollinearity. In this study, Bonferroni corrected P value was used for significance threshold. We found 2 metabolites with overlaps, which are lipids. Docosatrienoate (22:3n3) was found to be causally associated with a decreased risk of EC, as evidenced by the EC GWAS data (from Jiang et al) (odds ratio [OR] = 0.620, 95% confidence interval [CI] = 0.390-0.986, P = .044) and the EC GWAS data (from FINNGEN) (OR = 0.77, 95% CI = 0.6-0.99, P = .042), these results were consistent across both data sets. Another overlapping metabolite, glycosyl-N-(2-hydroxyneuramoyl)-sphingosine, was associated with the risk of ES, with EC GWAS data (from Jiang L et al) (OR = 1.536, 95% CI = 1.000-2.360, P = .049), while EC GWAS data (from FINNGEN) (OR = 0.733, 95% CI = 0.574-0.937, P = .013), the 2 data had opposite conclusions. The findings of this study indicate a potential association between lipid metabolites (Docosatrienoate (22:3n3) and glycosyl-N-(2-hydroxynervonoyl)-sphingosine (d18:1/24:1 (2OH))) and the risk of esophageal carcinogenesis.
Collapse
Affiliation(s)
- Jieyin Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Silin Wu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
- School of Clinical Medicine, North Sichuan Medical College, Sichuan, China
| | - Ye Huang
- Department of Nursing, Nursing School, Chengdu Medical College, Chengdu, China
| | - Yi Deng
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| | - Ke Yu
- Department of General Medical Practice, General Hospital of PLA Western Theater Command, Chengdu, China
| |
Collapse
|
9
|
Balstad TR, Bråtveit M, Solheim TS, Koteng LH, Bye A, Jakobsen RD, Schødt-Osmo B, Fjeldstad SH, Erichsen M, Vagnildhaug OM, Paur I, Ottestad I. Validity of dietary intake methods in cancer cachexia. Curr Opin Support Palliat Care 2024; 18:145-153. [PMID: 38980805 DOI: 10.1097/spc.0000000000000709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
PURPOSE OF REVIEW Accurate assessment of dietary intake, especially energy and protein intake, is crucial for optimizing nutritional care and outcomes in patients with cancer. Validation of dietary assessment methods is necessary to ensure accuracy, but the validity of these methods in patients with cancer, and especially in those with cancer cachexia, is uncertain. Validating nutritional intake is complex because of the variety of dietary methods, lack of a gold standard method, and diverse validation measures. Here, we review the literature on validations of dietary intake methods in patients with cancer, including those with cachexia, and highlight the gap between current validation efforts and the need for accurate dietary assessment methods in this population. RECENT FINDINGS We analyzed eight studies involving 1479 patients with cancer to evaluate the accuracy and reliability of 24-hour recalls, food records, and food frequency questionnaires in estimating energy and protein intake. We discuss validation methods, including comparison with biomarkers, indirect calorimetry, and relative validation of dietary intake methods. SUMMARY Few have validated dietary intake methods against objective markers in patients with cancer. While food records and 24-hour recalls show potential accuracy for energy and protein intake, this may be compromised in hypermetabolic patients. Additionally, under- and overreporting of intake may be less frequent, and the reliability of urinary nitrogen as a protein intake marker in patients with cachexia needs further investigation. Accurate dietary assessment is important for enhancing nutritional care outcomes in cachexia trials, requiring validation at multiple time points throughout the cancer trajectory.
Collapse
Affiliation(s)
- Trude R Balstad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical Medicine, Clinical Nutrition Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Marianne Bråtveit
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tora S Solheim
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Cancer Clinic, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Lisa Heide Koteng
- European Palliative Care Research Centre (PRC), Department of Oncology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Asta Bye
- European Palliative Care Research Centre (PRC), Department of Oncology, Oslo University Hospital and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Nursing and Health Promotion, Faculty of Health Sciences, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Rasmus Dahl Jakobsen
- Department of Clinical Medicine, Clinical Nutrition Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Siv Hilde Fjeldstad
- Clinical Nutrition Center, University Hospital of North Norway, Tromsø, Norway
| | - Marianne Erichsen
- Department of Clinical Medicine, Clinical Nutrition Research Group, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ola Magne Vagnildhaug
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Cancer Clinic, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Ingvild Paur
- Department of Clinical Medicine, Clinical Nutrition Research Group, UiT The Arctic University of Norway, Tromsø, Norway
- Norwegian Advisory Unit on Disease-Related Undernutrition, and Section of Clinical Nutrition, Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Norway
| | - Inger Ottestad
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway
- The Clinical Nutrition Outpatient Clinic, Section of Clinical Nutrition, Department of Clinical Service, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
10
|
Ghoreishy SM, Zeraattalab-Motlagh S, Amiri Khosroshahi R, Hemmati A, Noormohammadi M, Mohammadi H. Dose-Dependent Impacts of Omega-3 Fatty Acids Supplementation on Anthropometric Variables in Patients With Cancer: Results From a Systematic Review and Meta-Analysis of Randomized Clinical Trials. Clin Nutr Res 2024; 13:186-200. [PMID: 39165286 PMCID: PMC11333147 DOI: 10.7762/cnr.2024.13.3.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/22/2024] Open
Abstract
Meta-analyses have been conducted with conflicting results on this topic. Due to missing several eligible studies in previous meta-analysis by Lam et al., we conducted an extensive systematic review and dose-response meta-analysis of randomized controlled trials in this regard. A comprehensive search was conducted across various databases, including MEDLINE/PubMed, ISI Web of Knowledge, Scopus, and Google Scholar, until November 2023. Based on the analysis of 33 studies comprising 2,047 individuals, it was found that there was a significant increase in body weight for each 1 g/day increase in omega-3 lipids (standardized MD [SMD], 0.52 kg; 95% confidence interval [CI], 0.31, 0.73; I2 = 95%; Grading of Recommendations Assessment, Development and Evaluation [GRADE] = low). Supplementation of omega-3 fatty acids did not yield a statistically significant impact on body mass index (BMI) (SMD, 0.12 kg/m2; 95% CI, -0.02, 0.27; I2 = 79%; GRADE = very low), lean body mass (LBM) (SMD, -0.02 kg; 95% CI, -0.43, 0.39; I2 = 97%; GRADE = very low), fat mass (SMD, 0.45 kg; 95% CI, -0.25, 1.15; I2 = 96%; GRADE = low), and body fat (SMD, 0.30%; 95% CI, -0.90, 1.51; I2 = 96%; GRADE = very low). After excluding 2 studies, the findings were significant for BMI. Regarding the results of the dose-response analysis, body weight increased proportionally by increasing the dose of omega-3 supplementation up to 4 g/day. Omega-3 fatty acid supplementation can improve body weight, but not BMI, LBM, fat mass, or body fat in cancer patients; large-scale randomized trials needed for more reliable results. Trial Registration PROSPERO Identifier: CRD42023395341.
Collapse
Affiliation(s)
- Seyed Mojtaba Ghoreishy
- Student Research Committee, School of Public Health, Iran University of Medical Sciences, Tehran 14665-354, Iran
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 14665-354, Iran
| | | | - Reza Amiri Khosroshahi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran 14155-6117, Iran
| | - Amirhossein Hemmati
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran 14155-6117, Iran
| | - Morvarid Noormohammadi
- Student Research Committee, School of Public Health, Iran University of Medical Sciences, Tehran 14665-354, Iran
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran 14665-354, Iran
| | - Hamed Mohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran 14155-6117, Iran
| |
Collapse
|
11
|
Mahé M, Seegers V, Vansteene D. Correlation between changes in nutritional status and tumor response in patients receiving immunotherapy for lung cancer (NUTIMMUNO study). Support Care Cancer 2024; 32:312. [PMID: 38676729 DOI: 10.1007/s00520-024-08519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
Malnutrition is a common condition in lung cancer, and it is an independent prognostic factor. The main objective of this study was to determine whether an early improvement at 3 months in the nutritional status (NS) of patients undergoing immune checkpoint inhibitor (ICI) is associated with a tumor response to treatment at 6-month follow-up. The clinical data of 106 patients initiating ICI for bronchopulmonary non-small cell lung cancer (NCSLC) were retrospectively reviewed. NS was defined according to the HAS 2019 recommendation, depending on BMI, percentage of weight loss, and albuminemia. NS was assessed at baseline (M0) and 3 months (M3) after ICI treatment initiation according to 3 categories: well-nourished, malnourished, and very malnourished. The NS evolution of the 92 patients who were still alive at 3 months was determined. The proportion of patients with malnutrition at M0 and M3 was 39.6% and 43.3%. Median follow-up was 18.7 months. OS and PFS were longer for patients in the M0 well-nourished group than in the malnourished and very malnourished groups. Patients who remained well-nourished had a significantly better ICI success rate at 6 months than patients who remained malnourished or improved or deteriorated their NS. OS was significantly longer for remaining well-nourished patients compared to the amelioration group and the degradation group. PFS was not significantly modified between the 4 evolution groups. Maintaining good NS during the first months of ICI treatment leads to better OS and objective response rate than remaining malnourished or early deteriorating NS. However, an early improvement in NS does not seem to predict a good tumor response to treatment and not a better OS either.
Collapse
Affiliation(s)
- Marie Mahé
- Institut de Cancérologie de L'Ouest, 49055, Angers, France.
| | | | - Damien Vansteene
- Institut de Cancérologie de L'Ouest, 44805, Saint Herblain, France
| |
Collapse
|
12
|
Lee CM, Kang JH, Go SI. Clinical implication of megestrol acetate in metastatic gastric cancer: a big data analysis from Health Insurance Review and Assessment (HIRA) database. Support Care Cancer 2024; 32:249. [PMID: 38530439 DOI: 10.1007/s00520-024-08430-5] [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: 08/07/2023] [Accepted: 03/08/2024] [Indexed: 03/28/2024]
Abstract
OBJECTIVE Megestrol acetate (MA) is used to manage anorexia and cachexia in patients with advanced cancer. This study investigated the prescription patterns of MA in patients with metastatic gastric cancer, as well as evaluated its impact on survival outcomes and the incidence of venous thromboembolism (VTE). METHODS A Health Insurance Review and Assessment (HIRA) service database was used to investigate differences in baseline characteristics, survival, and the incidence of VTE according to MA prescription patterns (i.e., prescription vs. no prescription) in patients diagnosed with metastatic gastric cancer from July 2014 to December 2015. RESULTS A total of 1938 patients were included in this study. In total, 65% of the patients were prescribed MA. Older age, treatment in tertiary hospitals, and palliative chemotherapy were statistically significant predictive factors for MA prescription. Continuous prescription of MA was observed in 37% of patients. There was no statistically significant difference in survival between the MA and non-MA prescription groups on multivariate analysis. Among the 1427 patients included in the analysis for VTE incidence, 4.3% and 2.9% were diagnosed with VTE during the follow-up period in the MA and non-MA prescription groups, respectively. However, there was no statistically significant difference in VTE diagnosis between the groups on multivariate analysis. CONCLUSION MA is commonly prescribed for metastatic gastric cancer, especially in elderly patients and those undergoing palliative chemotherapy, without significantly affecting survival or VTE risk.
Collapse
Affiliation(s)
- Chang Min Lee
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Jung Hun Kang
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Se-Il Go
- Division of Hematology/Oncology, Department of Internal Medicine, Institute of Health Science, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, 11 Samjeongja-Ro, Changwon, 51472, Republic of Korea.
| |
Collapse
|
13
|
Ruan GT, Deng L, Xie HL, Shi JY, Liu XY, Zheng X, Chen Y, Lin SQ, Zhang HY, Liu CA, Ge YZ, Song MM, Hu CL, Zhang XW, Yang M, Hu W, Cong MH, Zhu LC, Wang KH, Shi HP. Systemic inflammation and insulin resistance-related indicator predicts poor outcome in patients with cancer cachexia. Cancer Metab 2024; 12:3. [PMID: 38273418 PMCID: PMC10809764 DOI: 10.1186/s40170-024-00332-8] [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: 07/12/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The C-reactive protein (CRP)-triglyceride-glucose (TyG) index (CTI), which is a measure representing the level of inflammation and insulin resistance (IR), is related to poor cancer prognosis; however, the CTI has not been validated in patients with cancer cachexia. Thus, this study aimed to explore the potential clinical value of the CTI in patients with cancer cachexia. METHODS In this study, our prospective multicenter cohort included 1411 patients with cancer cachexia (mean age 59.45 ± 11.38, 63.3% male), which was a combined analysis of multiple cancer types. We randomly selected 30% of the patients for the internal test cohort (mean age 58.90 ± 11.22% 61.4% male). Additionally, we included 307 patients with cancer cachexia in the external validation cohort (mean age 61.16 ± 11, 58.5% male). Receiver operating characteristic (ROC) and calibration curves were performed to investigate the prognostic value of CTI. The prognostic value of the CTI was also investigated performing univariate and multivariate survival analyses. RESULTS The survival curve indicated that the CTI showed a significant prognostic value in the total, internal, and external validation cohorts. Prognostic ROC curves and calibration curves revealed that the CTI showed good consistency in predicting the survival of patients with cancer cachexia. Multivariate survival analysis showed that an elevated CTI increased the risk of death by 22% (total cohort, 95% confidence interval [CI] = 1.13-1.33), 34% (internal test cohort, 95%CI = 1.11-1.62), and 35% (external validation cohort, 95%CI = 1.14-1.59) for each increase in the standard deviation of CTI. High CTI reliably predicted shorter survival (total cohort, hazard ratio [HR] = 1.45, 95%CI = 1.22-1.71; internal test cohort, HR = 1.62, 95%CI = 1.12-2.36; external validation cohort, HR = 1.61, 95%CI = 1.15-2.26). High CTI significantly predicted shorter survival in different tumor subgroups, such as esophageal [HR = 2.11, 95%CI = 1.05-4.21] and colorectal cancer [HR = 2.29, 95%CI = 1.42-3.71]. The mediating effects analysis found that the mediating proportions of PGSGA, ECOG PS, and EORTC QLQ-C30 on the direct effects of CTI were 21.72%, 19.63%, and 11.61%, respectively We found that there was a significant positive correlation between the CTI and 90-day [HR = 2.48, 95%CI = 1.52-4.14] and 180-day mortality [HR = 1.77,95%CI = 1.24-2.55] in patients with cancer cachexia. CONCLUSION The CTI can predict the short- and long-term survival of patients with cancer cachexia and provide a useful prognostic tool for clinical practice.
Collapse
Affiliation(s)
- Guo-Tian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Li Deng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Hai-Lun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Jin-Yu Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xiao-Yue Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Yue Chen
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Shi-Qi Lin
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - He-Yang Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Chen-An Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Yi-Zhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Meng-Meng Song
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Chun-Lei Hu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xiao-Wei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Wen Hu
- Clinical Nutrition Department, Sichuan University West China Hospital, Chengdu, 610041, Sichuan, China
| | - Ming-Hua Cong
- Comprehensive Oncology Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100038, China
| | - Li-Chen Zhu
- Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, China
| | - Kun-Hua Wang
- Yunnan University, Kunming, 650091, China
- General Surgery Clinical Medical Center of Yunnan Province, Kunming, 650032, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China.
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China.
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China.
| |
Collapse
|
14
|
Alvarez-Altamirano K, Miramontes-Balcon K, Cárcoba-Tenorio C, Bejarano-Rosales M, Amanda-Casillas M, Serrano-Olvera JA, Fuchs-Tarlovsky V. Resting energy expenditure changes after antineoplastic treatment in gynecological cancer: a prospective pilot study. NUTR HOSP 2023; 40:1199-1206. [PMID: 37929857 DOI: 10.20960/nh.04768] [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: 11/07/2023] Open
Abstract
Introduction Introduction: energy metabolism in cancer patients is influenced by different factors. However, the effect of antineoplastic treatment is not clear, especially in women. Objective: to evaluate resting energy expenditure (REE) by indirect calorimetry (IC) before (T0) and after (T1) first cycle period of antineoplastic therapy: radiotherapy (RT), chemotherapy (CT), and concomitant chemoradiation therapy (CRT), quality of life (QoL) and accuracy of REE were compared with international guidelines recommendations per kilogram (European Society for Clinical Nutrition and Metabolism [ESPEN]). Methods: an observational, longitudinal study was conducted in women with gynecological cancer diagnosis undergoing antineoplastic treatment: RT, CT and CRT. Weight loss, actual body weight and height were measured. REE was evaluated in T0-T1 and compared with ESPEN recommendations. Kruskal-Wallis test and Bland-Alman analysis were used to determine the agreement (± 10 % of energy predicted) of REE adjusted by physical activity (TEE) compared with ESPEN recommendations, respectively. Results: fifty-four women with cancer were included: 31.5 % (n = 17) for RT group, 31.5 % (n = 17) for CT group and 37 % (n = 20) for CRT group. REE showed statistical differences between T0 and T1 in the total population (p = 0.018), but these were not associated with anticancer therapy groups (p > 0.05). QoL had no significant changes after treatment (p > 0.05). Accuracy of 25 and 30 kcal/kg compared to TEE was less than 30 %. Conclusion: REE in women with gynecological cancer decreased after antineoplastic treatments but this is not associated with a particular antineoplastic therapy. It is needed to develop research to determine the accuracy of ESPEN recommendations with TEE estimated by IC and clinical factors in women with cancer.
Collapse
|
15
|
Dave S, Patel BM. Deliberation on debilitating condition of cancer cachexia: Skeletal muscle wasting. Fundam Clin Pharmacol 2023; 37:1079-1091. [PMID: 37474262 DOI: 10.1111/fcp.12931] [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: 01/30/2023] [Revised: 05/18/2023] [Accepted: 06/08/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Cancer cachexia is a debilitating syndrome associated with marked body loss because of muscular atrophy and fat loss. There are several mechanisms contributing to the pathogenesis of cachexia. The presence of the tumor releases cytokines from inflammatory and immune cells, which play a significant role in activating and deactivating certain pathways associated with protein, carbohydrate, and lipid metabolism. This review focuses on various cascades involving an imbalance between protein synthesis and degradation in the skeletal muscles. OBJECTIVES This study aimed to elucidate the mechanisms involved in skeletal muscle wasting phenomenon over the last few years. METHODS This article briefly overviews different pathways responsible for muscle atrophy in cancer cachexia. Studies published up to April 2023 were included. Important findings and study contributions were chosen and compiled using several databases including PubMed, Google Scholar, Science Direct, and ClinicalTrials.gov using relevant keywords. RESULTS Cancer cachexia is a complex disease involving multiple factors resulting in atrophy of skeletal muscles. Systemic inflammation, altered energy balance and carbohydrate metabolism, altered lipid and protein metabolism, and adipose tissue browning are some of the major culprits in cancer cachexia. Increased protein degradation and decreased protein synthesis lead to muscle atrophy. Changes in signaling pathway like ubiquitin-proteasome, autophagy, mTOR, AMPK, and IGF-1 also lead to muscle wasting. Physical exercise, nutritional supplementation, steroids, myostatin inhibitors, and interventions targeting on inflammation have been investigated to treat cancer cachexia. Some therapy showed positive results in preclinical and clinical settings, although more research on the efficacy and safety of the treatment should be done. CONCLUSION Muscle atrophy in cancer cachexia is the result of multiple complex mechanisms; as a result, a lot more research has been done to describe the pathophysiology of the disease. Targeted therapy and multimodal interventions can improve clinical outcomes for patients.
Collapse
Affiliation(s)
- Srusti Dave
- Institute of Pharmacy, Nirma University, Ahmedabad, Gujarat, India
| | - Bhoomika M Patel
- School of Medico-legal Studies, National Forensic Sciences University, Gandhinagar, India
| |
Collapse
|
16
|
Hanna L, Porter J, Bauer J, Nguo K. Energy Expenditure in Upper Gastrointestinal Cancers: a Scoping Review. Adv Nutr 2023; 14:1307-1325. [PMID: 37562709 PMCID: PMC10721480 DOI: 10.1016/j.advnut.2023.08.002] [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: 04/17/2023] [Revised: 07/18/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
Malnutrition is prevalent in people with upper gastrointestinal (GI) cancers and is associated with shorter survival and poor quality of life. In order to effectively prevent or treat malnutrition, nutrition interventions must ensure appropriate energy provision to meet daily metabolic demands. In practice, the energy needs of people with cancer are frequently estimated from predictive equations which are not cancer-specific and are demonstrated to be inaccurate in this population. The purpose of this scoping review was to synthesize the existing evidence regarding energy expenditure in people with upper GI cancer. Three databases (Ovid MEDLINE, Embase via Ovid, CINAHL plus) were systematically searched to identify studies reporting on resting energy expenditure using indirect calorimetry and total energy expenditure using doubly labeled water (DLW) in adults with any stage of upper GI cancer at any point from diagnosis. A total of 57 original research studies involving 2,125 individuals with cancer of the esophagus, stomach, pancreas, biliary tract, or liver were eligible for inclusion. All studies used indirect calorimetry, and one study used DLW to measure energy expenditure, which was reported unadjusted in 42 studies, adjusted for body weight in 32 studies, and adjusted for fat-free mass in 13 studies. Energy expenditure in upper GI cancer was compared with noncancer controls in 19 studies and measured compared with predicted energy expenditure reported in 31 studies. There was heterogeneity in study design and in reporting of important clinical characteristics between studies. There was also substantial variation in energy expenditure between studies and within and between cancer types. Given this heterogeneity and known inaccuracies of predictive equations in patients with cancer, energy expenditure should be measured in practice wherever feasible. Additional research in cohorts defined by cancer type, stage, and treatment is needed to further characterize energy expenditure in upper GI cancer.
Collapse
Affiliation(s)
- Lauren Hanna
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Victoria, Australia; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia.
| | - Judi Porter
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Victoria, Australia; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Judy Bauer
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Victoria, Australia
| | - Kay Nguo
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
17
|
López-Plaza B, Gil Á, Menéndez-Rey A, Bensadon-Naeder L, Hummel T, Feliú-Batlle J, Palma-Milla S. Effect of Regular Consumption of a Miraculin-Based Food Supplement on Taste Perception and Nutritional Status in Malnourished Cancer Patients: A Triple-Blind, Randomized, Placebo-Controlled Clinical Trial-CLINMIR Pilot Protocol. Nutrients 2023; 15:4639. [PMID: 37960292 PMCID: PMC10648678 DOI: 10.3390/nu15214639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Taste disorders are common among cancer patients undergoing chemotherapy, with a prevalence ranging from 20% to 86%, persisting throughout treatment. This condition leads to reduced food consumption, increasing the risk of malnutrition. Malnutrition is associated not only with worse treatment efficacy and poor disease prognosis but also with reduced functional status and quality of life. The fruit of Synsepalum dulcificum (Daniell), commonly known as miracle berry or miracle fruit, contains miraculin, a taste-modifying protein with profound effects on taste perception. The CLINMIR Protocol is a triple-blind, randomized, placebo-controlled clinical trial designed to evaluate the regular consumption of a food supplement containing a miraculin-based novel food, dried miracle berry (DMB), on the taste perception (measured through electrogustometry) and nutritional status (evaluated through the GLIM Criteria) of malnourished cancer patients under active antineoplastic treatment. To this end, a pilot study was designed with 30 randomized patients divided into three study arms (150 mg DMB + 150 mg freeze-dried strawberries, 300 mg DMB, or placebo) for three months. Throughout the five main visits, an exhaustive assessment of different parameters susceptible to improvement through regular consumption of the miraculin-based food supplement will be conducted, including electrical and chemical taste perception, smell perception, nutritional and morphofunctional assessment, diet, quality of life, the fatty acid profile of erythrocytes, levels of inflammatory and cancer-associated cytokines, oxidative stress, antioxidant defense system, plasma metabolomics, and saliva and stool microbiota. The primary anticipated result is that malnourished cancer patients with taste distortion who consume the miraculin-based food supplement will report an improvement in food taste perception. This improvement translates into increased food intake, thereby ameliorating their nutritional status and mitigating associated risks. Additionally, the study aims to pinpoint the optimal dosage that provides maximal benefits. The protocol adheres to the SPIRIT 2013 Statement, which provides evidence-based recommendations and is widely endorsed as an international standard for trial protocols. The clinical trial protocol has been registered at the platform for Clinical Trials (NCT05486260).
Collapse
Affiliation(s)
- Bricia López-Plaza
- Nutrition Research Group, La Paz University Hospital Institute for Health Research (IdiPAZ), 28046 Madrid, Spain;
- Medicine Department, Faculty of Medicine, Complutense University of Madrid, Plaza de Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n, Armilla, 18016 Granada, Spain
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | | | - Thomas Hummel
- Smell & Taste Clinic, Department of Otorhinolaryngology, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany;
| | - Jaime Feliú-Batlle
- Oncology Department, Hospital La Paz Institute for Health Research—IdiPAZ, Hospital Universitario La Paz, 28029 Madrid, Spain;
- CIBERONC (CIBER Cancer), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Medicine Department, Faculty of Medicine, Autonomous University of Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain;
| | - Samara Palma-Milla
- Medicine Department, Faculty of Medicine, Autonomous University of Madrid, Arzobispo Morcillo 4, 28029 Madrid, Spain;
- Nutrition Department, Hospital University La Paz, 28046 Madrid, Spain
| |
Collapse
|
18
|
Ling T, Liu J, Dong L, Liu J. The roles of P-selectin in cancer cachexia. Med Oncol 2023; 40:338. [PMID: 37870739 DOI: 10.1007/s12032-023-02207-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/30/2023] [Indexed: 10/24/2023]
Abstract
P-selectin, a cell adhesion molecule of the selectin family, is expressed on the surface of activated endothelial cells (ECs) and platelets. Binding of P-selectin to P-selectin glycoprotein ligand-1 (PSGL-1) supports the leukocytes capture and rolling on stimulated ECs and increases the aggregation of leukocytes and activated platelets. Cancer cachexia is a systemic inflammation disorder characterized by metabolic disturbances, reduced body weight, loss of appetite, fat depletion, and progressive muscle atrophy. Cachexia status is associated with increased pro-inflammatory cytokines such as tumor necrosis factor-alpha (TNF-α), which activates ECs to release P-selectin. Single-nucleotide polymorphisms (SNPs) loci of P-selectin encoding gene SELP are associated with higher level of plasma P-selectin and increase the susceptibility to cachexia in cancer patients. Elevated P-selectin expression has been observed in the hypothalamus, liver, and gastrocnemius muscle in animal models with cancer cachexia. Increased P-selectin may cause excessive inflammatory processes, muscle atrophy, and blood hypercoagulation, thus facilitating the development of cancer cachexia. In this review, physiological functions of P-selectin and its potential roles in cancer cachexia have been summarized. We also discuss the therapeutic potential of P-selectin inhibitors for the treatment of cancer cachexia.
Collapse
Affiliation(s)
- Tingting Ling
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, School of Clinical Medicine, Weifang Medical College, Weifang, 261053, China
| | - Jing Liu
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, 250014, China
| | - Liang Dong
- Department of Respiratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Ju Liu
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, 250014, China.
| |
Collapse
|
19
|
Vedire Y, Nitsche L, Tiadjeri M, McCutcheon V, Hall J, Barbi J, Yendamuri S, Ray AD. Skeletal muscle index is associated with long term outcomes after lobectomy for non-small cell lung cancer. BMC Cancer 2023; 23:778. [PMID: 37598139 PMCID: PMC10439565 DOI: 10.1186/s12885-023-11210-9] [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: 11/22/2022] [Accepted: 07/23/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Skeletal muscle indices have been associated with improved peri-operative outcomes after surgical resection of non-small-cell lung cancer (NSCLC). However, it is unclear if these indices can predict long term cancer specific outcomes. METHODS NSCLC patients undergoing lobectomy at our institute between 2009-2015 were included in this analysis (N = 492). Preoperative CT scans were used to quantify skeletal muscle index (SMI) at L4 using sliceOmatic software. Cox proportional modelling was performed for overall (OS) and recurrence free survival (RFS). RESULTS For all patients, median SMI was 45.7 cm2/m2 (IQR, 40-53.8). SMI was negatively associated with age (R = -0.2; p < 0.05) and positively associated with BMI (R = 0.46; P < 0.05). No association with either OS or RFS was seen with univariate cox modelling. However, multivariable modelling for SMI with patient age, gender, race, smoking status, DLCO and FEV1 (% predicted), American Society of Anesthesiology (ASA) score, tumor histology and stage, and postoperative neoadjuvant therapy showed improved OS (HR = 0.97; P = 0.0005) and RFS (HR = 0.97; P = 0.01) with SMI. Using sex specific median SMI as cutoff, a lower SMI was associated with poor OS (HR = 1.65, P = 0.001) and RFS (HR = 1.47, P = 0.03). CONCLUSIONS SMI is associated with improved outcomes after resection of NSCLC. Further studies are needed to understand the biological basis of this observation. This study provides additional rationale for designing and implementation of rehabilitation trials after surgical resection, to gain durable oncologic benefit.
Collapse
Affiliation(s)
- Yeshwanth Vedire
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Lindsay Nitsche
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Madeline Tiadjeri
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Victor McCutcheon
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Jack Hall
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
- Department of Physical Therapy and Rehabilitation, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Joseph Barbi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, Ny, 14263, USA
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| | - Andrew D Ray
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
- Department of Rehabilitation, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
20
|
Ford KL, Pichard C, Sawyer MB, Trottier CF, Disi IR, Purcell SA, Ghosh S, Siervo M, Deutz NE, Prado CM. Total energy expenditure assessed by 24-h whole-room indirect calorimeter in patients with colorectal cancer: baseline findings from the PRIMe study. Am J Clin Nutr 2023; 118:422-432. [PMID: 37290740 DOI: 10.1016/j.ajcnut.2023.06.002] [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: 01/03/2023] [Revised: 05/19/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Total energy expenditure (TEE) determines energy requirements, but objective data in patients with cancer are limited. OBJECTIVES We aimed to characterize TEE, investigate its predictors, and compare TEE with cancer-specific predicted energy requirements. METHODS This cross-sectional analysis included patients with stages II-IV colorectal cancer from the Protein Recommendation to Increase Muscle (PRIMe) trial. TEE was assessed by 24-h stay in a whole-room indirect calorimeter before dietary intervention and compared with cancer-specific predicted energy requirements (25-30 kcal/kg). Generalized linear models, paired-samples t tests, and Pearson correlation were applied. RESULTS Thirty-one patients (56 ± 10 y; body mass index [BMI]: 27.9 ± 5.5 kg/m2; 68% male) were included. Absolute TEE was higher in males (mean difference: 391 kcal/d; 95% CI: 167, 616 kcal/d; P < 0.001), patients with colon cancer (mean difference: 279 kcal/d; 95% CI: 73, 485 kcal/d; P = 0.010), and patients with obesity (mean difference: 393 kcal/d; 95% CI: 182, 604 kcal/d; P < 0.001). Appendicular lean soft tissue (β: 46.72; 95% CI: 34.27, 59.17; P < 0.001) and tumor location (colon-β: 139.69; 95% CI: 19.44, 259.95; P = 0.023) independently predicted TEE when adjusted for sex. Error between measured TEE and energy requirements predicted by 25 kcal/kg (mean difference: 241 kcal/d; 95% CI: 76, 405 kcal/d; P = 0.010) or 30 kcal/kg (mean difference: 367 kcal/d; 95% CI: 163, 571 kcal/d; P < 0.001) was higher for patients with obesity, and proportional error was observed (25 kcal/kg: r = -0.587; P < 0.001; and 30 kcal/kg: r = -0.751; P < 0.001). TEE (mean difference: 25 kcal/kg; 95% CI: 24, 27 kcal/kg) was below predicted requirements using 30 kcal/kg (-430 ± 322 kcal/d; P < 0.001). CONCLUSIONS This is the largest study to assess TEE of patients with cancer using whole-room indirect calorimeter and highlights the need for improved assessment of energy requirements in this population. Energy requirements predicted using 30 kcal/kg overestimated TEE by 1.44 times in a controlled sedentary environment and TEE was outside of the predicted requirement range for most. Special considerations are warranted when determining TEE of patients with colorectal cancer, such as BMI, body composition, and tumor location. This is a baseline cross-sectional analysis from a clinical trial registered at clinicaltrials.gov as NCT02788955 (https://clinicaltrials.gov/ct2/show/NCT02788955).
Collapse
Affiliation(s)
- Katherine L Ford
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada
| | - Claude Pichard
- Clinical Nutrition, Geneva University Hospital, Geneva, Switzerland
| | | | - Claire F Trottier
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada
| | - Ilana Roitman Disi
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada; Department of Anesthesia, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah A Purcell
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada; Department of Medicine, University of British Columbia, Vancouver, Canada; Department of Biology, University of British Columbia Okanagan, Kelowna, Canada
| | - Sunita Ghosh
- Department of Oncology, University of Alberta, Edmonton, Canada
| | - Mario Siervo
- School of Population Health, Curtin University, Perth, Australia; Curtin Dementia Centre of Excellence, enAble Institute, Curtin University, Perth, Australia
| | - Nicolaas Ep Deutz
- Center for Translational Research in Aging and Longevity, Texas A&M University, College Station, United States
| | - Carla M Prado
- Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada.
| |
Collapse
|
21
|
De Moura A, Turpin A, Neuzillet C. [Nutritional supportive care in the course of patients with esophagogastric cancers]. Bull Cancer 2023; 110:540-551. [PMID: 36202638 DOI: 10.1016/j.bulcan.2022.08.007] [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: 05/02/2022] [Revised: 08/02/2022] [Accepted: 08/13/2022] [Indexed: 05/09/2023]
Abstract
Supportive care plays a central role in the management of patients with esophagogastric cancers, at all disease stages. Malnutrition has a high prevalence in this population, reaching up to 60 % of the patients. Sarcopenia and cachexia are also common. These complications have negative impact on functional abilities, quality of life and overall survival. They impair anti-tumor treatments effectiveness and increase their toxicity. Early detection and management are needed, before reaching advanced stages, which are refractory to therapeutic interventions. Specific nutritional support is recommended, relying on different nutritional support tools (dietetic counseling, oral supplements, artificial nutrition), depending on the clinical situation. When artificial nutrition is recommended, enteral nutrition (nasogastric tube, gastrostomy or jejunostomy) should be preferred. When enteral nutrition is impossible or insufficient, parenteral nutrition could be necessary. For patients with advanced esophagogastric cancers, digestive prostheses and decompressive radiation therapy may have a symptomatic benefit on dysphagia. Adapted physical activity is also recommended at all stages of cancer care and ongoing clinical trials will help to specify its modalities and to optimize its place in the therapeutic strategy. Finally, psychosocial support could be useful. A combined approach of these different interventions on the nutritional, physical and psychological aspects is beneficial for patients with esophagogastric cancers. This multimodal and multidisciplinary approach applies to both the early stages of the disease, with prehabilitation and/or rehabilitation to reduce perioperative morbidity and mortality and the advanced stages, with a benefit on survival and quality of life, in parallel with anti-tumor treatments.
Collapse
Affiliation(s)
- Alexandre De Moura
- Institut Curie, université Versailles Saint-Quentin, université Paris Saclay, département d'oncologie médicale, 35, rue Dailly, 92210 Saint-Cloud, France.
| | - Anthony Turpin
- Université de Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277, CANTHER, Cancer heterogeneity plasticity and resistance to therapies, 59000 Lille, France
| | - Cindy Neuzillet
- Institut Curie, université Versailles Saint-Quentin, université Paris Saclay, département d'oncologie médicale, 35, rue Dailly, 92210 Saint-Cloud, France
| |
Collapse
|
22
|
Clinical significance of the preoperative prognostic nutritional index on age/comorbidity burdens in patients with resectable non-small cell lung cancer. Surg Today 2023; 53:681-691. [PMID: 36720742 DOI: 10.1007/s00595-023-02650-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/18/2022] [Indexed: 02/02/2023]
Abstract
PURPOSE Objective nutritional scoring systems using preoperative blood samples have shown the potential to predict the postoperative outcomes of patients with non-small cell lung cancer (NSCLC). However, it remains unclear whether the prognostic impact depends on age and comorbid burdens. We conducted this study to validate the impact of preoperative nutritional status, stratified with age and comorbidity. METHODS We reviewed the preoperative prognostic nutritional index (PNI) and postoperative outcomes of 713 consecutive patients with completely resected NSCLC. RESULTS We identified the optimal cutoff values of the PNI as 46. Significantly higher postoperative complication rates and worse survival rates were observed in the low PNI (≤ 46) group, regardless of age/comorbidity burdens. Multivariate analysis showed that a low PNI (≤ 46) was an independent prognostic factor for poor overall survival (hazard ratio: 2.5). A matched-pair analysis gave consistent results, showing that a low PNI (≤ 46) was an independent prognostic factor for poor overall survival (OS; hazard ratio: 1.8) and recurrence-free survival (RFS; hazard ratio: 1.6). CONCLUSION Nutritional status, indexed by the PNI, is a strong prognostic factor for the postoperative outcomes of patients undergoing curative resection for NSCL, regardless of age/comorbidity burdens.
Collapse
|
23
|
Eysenbach G, Goldsack JC, Cordovano G, Downing A, Fields KK, Geoghegan C, Grewal U, Nieva J, Patel N, Rollison DE, Sah A, Said M, Van De Keere I, Way A, Wolff-Hughes DL, Wood WA, Robinson EJ. Advancing Digital Health Innovation in Oncology: Priorities for High-Value Digital Transformation in Cancer Care. J Med Internet Res 2023; 25:e43404. [PMID: 36598811 PMCID: PMC9850283 DOI: 10.2196/43404] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/17/2022] [Accepted: 11/30/2022] [Indexed: 01/05/2023] Open
Abstract
Although health care delivery is becoming increasingly digitized, driven by the pursuit of improved access, equity, efficiency, and effectiveness, progress does not appear to be equally distributed across therapeutic areas. Oncology is renowned for leading innovation in research and in care; digital pathology, digital radiology, real-world data, next-generation sequencing, patient-reported outcomes, and precision approaches driven by complex data and biomarkers are hallmarks of the field. However, remote patient monitoring, decentralized approaches to care and research, "hospital at home," and machine learning techniques have yet to be broadly deployed to improve cancer care. In response, the Digital Medicine Society and Moffitt Cancer Center convened a multistakeholder roundtable discussion to bring together leading experts in cancer care and digital innovation. This viewpoint highlights the findings from these discussions, in which experts agreed that digital innovation is lagging in oncology relative to other therapeutic areas. It reports that this lag is most likely attributed to poor articulation of the challenges in cancer care and research best suited to digital solutions, lack of incentives and support, and missing standardized infrastructure to implement digital innovations. It concludes with suggestions for actions needed to bring the promise of digitization to cancer care to improve lives.
Collapse
Affiliation(s)
| | | | | | | | - Karen K Fields
- Center for Digital Health, Moffitt Cancer Center, Tampa, FL, United States
| | | | | | - Jorge Nieva
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Nikunj Patel
- AstraZeneca PLC, Gaithersburg, MD, United States
| | - Dana E Rollison
- Center for Digital Health, Moffitt Cancer Center, Tampa, FL, United States
| | - Archana Sah
- AS Pharma Advisors, Inc, San Francisco, CA, United States
| | - Maya Said
- Outcomes4Me Inc, Boston, MA, United States
| | | | - Amanda Way
- Jazz Venture Partners, San Francisco, CA, United States
| | - Dana L Wolff-Hughes
- Division of Cancer Control and Populations Sciences, National Cancer Institute, Bethesda, MD, United States
| | - William A Wood
- Lineberger Comprehensive Cancer Center, University of North Carolina Chapel Hill, Chapel Hill, NC, United States
| | - Edmondo J Robinson
- Center for Digital Health, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
24
|
Tarachandani A, Karahanoglu FI, Messere A, Tarasenko L, LaRonde-Richard AM, Kessler N, Rossulek M, Plate H, Mahoney K, Santamaria M. Patient Willingness to Use Digital Health Technologies: A Quantitative and Qualitative Survey in Patients with Cancer Cachexia. Patient Prefer Adherence 2023; 17:1143-1157. [PMID: 37139257 PMCID: PMC10150793 DOI: 10.2147/ppa.s396347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 04/01/2023] [Indexed: 05/05/2023] Open
Abstract
Purpose The objective of this study was to gain insights into the patients' perspectives on the impact of cancer cachexia on physical activity and their willingness to wear digital health technology (DHT) devices in clinical trials. Patients and Methods We administered a quantitative 20-minute online survey on aspects of physical activity (on a 0-100 scale) to 50 patients with cancer cachexia recruited through Rare Patient Voice, LLC. A subset of 10 patients took part in qualitative 45-minute web-based interviews with a demonstration of DHT devices. Survey questions related to the impact of weight loss (a key characteristic in Fearon's cachexia definition) on physical activity, patients' expectations regarding desired improvements and their level of meaningful activities, as well as preferences for DHT. Results Seventy-eight percent of patients reported that their physical activity was impacted by cachexia, and for 77% of them, such impact was consistent over time. Patients perceived most impact of weight loss on walking distance, time and speed, and on level of activity during the day. Sleep, activity level, walking quality and distance were identified as the most meaningful activities to improve. Patients would like to see a moderate improvement of activity levels and consider it meaningful to perform physical activity of moderate intensity (eg, walk at normal pace) on a regular basis. The wrist was the preferred location for wearing a DHT device, followed by arm, ankle, and waist. Conclusion Most patients reported physical activity limitations since the occurrence of weight loss compatible with cancer-associated cachexia. Walking distance, sleep and quality of walk were the most meaningful activities to moderately improve, and patients consider moderate physical activity as meaningful. Finally, this study population found the proposed wear of DHT devices on the wrist and around the waist acceptable for the duration of clinical studies.
Collapse
Affiliation(s)
| | | | - Andrew Messere
- Early Clinical Development, Pfizer Inc., Cambridge, MA, USA
| | | | | | - Nancy Kessler
- Business Analytics and Insights, Pfizer Inc., New York, NY, USA
| | | | | | | | - Mar Santamaria
- Early Clinical Development, Pfizer Inc., Cambridge, MA, USA
- Correspondence: Mar Santamaria, Early Clinical Development, Pfizer Inc., 610 Main Street, Cambridge, MA, 02139, USA, Tel +1 617 852 5637, Fax +1 845 474 5357, Email
| |
Collapse
|
25
|
Huo Z, Chong F, Yin L, Li N, Zhang M, Guo J, Lin X, Fan Y, Zhang L, Zhang H, Shi M, He X, Lu Z, Liu J, Li W, Shi H, Xu H. Development and validation of an online dynamic nomogram system for predicting cancer cachexia among inpatients: a real-world cohort study in China. Support Care Cancer 2022; 31:72. [PMID: 36543973 DOI: 10.1007/s00520-022-07540-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Early recognition of cachexia is essential for ensuring the prompt intervention and treatment of cancer patients. However, the diagnosis of cancer cachexia (CC) usually is delayed. This study aimed to establish an accurate and high-efficiency diagnostic system for CC. METHODS A total of 4834 cancer inpatients were enrolled in the INSCOC project from July 2013 to June 2020. All cancer patients in the study were randomly assigned to a development cohort (n=3384, 70%) and a validation cohort (n=1450, 30%). The least absolute shrinkage and selection operator (LASSO) method and multivariable logistic regression were used to identify the independent predictors for developing the dynamic nomogram. Discrimination and calibration were adopted to evaluate the ability of nomogram. A decision curve analysis (DCA) was used to evaluate clinical use. RESULTS We combined 5 independent predictive factors (age, NRS2002, PG-SGA, QOL by the QLQ-C30, and cancer categories) to establish the online dynamic nomogram system. The C-index, sensitivity, and specificity of the nomo-system to predict CC was 0.925 (95%CI, 0.916-0.934, P < 0.001), 0.826, and 0.862 in the development set, while the values were 0.923 (95%CI, 0.909-0.937, P < 0.001), 0.854, and 0.829 in the validation set. In addition, the calibration curves of the diagnostic nomogram also presented good agreement with the actual situation. DCA showed that the model is clinically useful and can increase the clinical benefit in cancer patients. CONCLUSIONS This study developed an online dynamic nomogram system with outstanding accuracy to help clinicians and dieticians estimate the probability of cachexia. This simple-to-use online nomogram can increase the clinical benefit in cancer patients and is expected to be widely adopted.
Collapse
Affiliation(s)
- Zhenyu Huo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Feifei Chong
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Liangyu Yin
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Na Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Mengyuan Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jing Guo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Xin Lin
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Yang Fan
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Ling Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Hongmei Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Muli Shi
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Xiumei He
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Zongliang Lu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Jie Liu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China
| | - Wei Li
- Cancer Center of the First Affiliated Hospital of Jilin University, Changchun, 130021, China
| | - Hanping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
- Key Laboratory of Cancer FSMP for State Market Regulation, Beijing, 100038, China
| | - Hongxia Xu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, 400042, China.
| |
Collapse
|
26
|
Delpino FM, Figueiredo LM. Effects of omega-3 supplementation on lean body mass in cancer patients: a systematic review and meta-analysis. Eur J Clin Nutr 2022; 76:1636-1645. [PMID: 35173292 DOI: 10.1038/s41430-022-01100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 11/09/2022]
Abstract
Omega-3 fatty acids are bioactive nutrients with the potential to preserve lean body mass in individuals with cancer. This study aimed to review the literature on randomized clinical trials that evaluated the effects of omega-3 supplementation on lean body mass in cancer patients. As secondary objectives, we evaluated the effects of omega-3 supplementation on body mass index (BMI) and body weight. We conducted a systematic review and meta-analysis in the following databases: Pubmed, LILACS, Scielo, Scopus, Web of Science, Cochrane, and Embase. It included randomized clinical trials that investigated the effects of omega-3 supplementation on lean body mass in cancer patients. Observational studies, animal experiments, studies carried out with healthy humans, and non-randomized clinical trials were excluded. We utilized the Cochrane scale to assess the quality of the studies. A meta-analysis was carried out to evaluate the effect of omega-3 on lean body mass, BMI, and body weight. Fourteen studies were included, of which four showed significant results from omega-3 supplementation for lean body mass. In the meta-analysis, omega-3 fatty acids increased lean body mass by 0.17 kg compared to placebo, but without significant differences between the groups [SMD: 0.17; CI 95%: -0.01, 0.35; I2 = 41%]. For body weight, omega-3 showed a statistically significant effect [SMD: 0.26; CI 95%: 0.06, 0.45; I2 = 46%], whereas for BMI the results were not significant. This systematic review and meta-analysis showed no statistically significant effect from omega-3 on lean body mass and BMI. On the other hand, there was a statistical significance for body weight.
Collapse
Affiliation(s)
- Felipe Mendes Delpino
- Postgraduate Program in Nursing, Federal University of Pelotas, Rio Grande do Sul, Pelotas, Brazil.
- Faculty of Nursing, Federal University of Pelotas, Pelotas, Brazil.
| | | |
Collapse
|
27
|
Jin X, Xu XT, Tian MX, Dai Z. Omega-3 polyunsaterated fatty acids improve quality of life and survival, but not body weight in cancer cachexia: A systematic review and meta-analysis of controlled trials. Nutr Res 2022; 107:165-178. [PMID: 36283229 DOI: 10.1016/j.nutres.2022.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/17/2022] [Accepted: 09/20/2022] [Indexed: 12/27/2022]
Abstract
Several clinical trials have reported that patients with cancer cachexia can benefit from n-3 polyunsaturated fatty acids (n-3 PUFAs) supplements; however, the results have been conflicting. This systematic review and meta-analysis aimed to evaluate the effect of n-3 PUFAs on cancer cachexia. A search of the PubMed, Embase, and Cochrane Library databases was performed to identify the included randomized controlled trials. Trials including patients with cancer cachexia who were administered a course of n-3 PUFAs were included. A meta-analysis on body weight, lean body weight, proinflammatory factors, quality of life, and median duration of survival was conducted. A total of 12 randomized controlled trials with 1184 patients were included. No effect on body weight (standard mean difference [SMD], 0.10; 95% CI, -0.06 to 0.26; P = .236), lean body weight (SMD, -0.17; 95% CI, -0.36 to 0.03, P = .095), or proinflammatory factors (interleukin-6: SMD, 0.31; 95% CI, -0.14 to 0.75; P = .18; tumor necrosis factor-α: SMD, -0.85; 95% CI, -2.39 to 0.69; P = .28) was observed. The use of n-3 PUFAs was associated with a significant improvement in quality of life (SMD, 0.70; 95% CI, 0.01-1.40; P = .048) and median duration of survival (median survival ratio, 1.10; 95% CI, 1.02-1.19; P = .014). For patients with cancer cachexia, our meta-analysis indicated that n-3 PUFAs improved quality of life and survival, but not body weight.
Collapse
Affiliation(s)
- Xin Jin
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xin-Tian Xu
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng-Xing Tian
- Department of Clinical Nutrition, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhu Dai
- Department of Pharmacy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
28
|
Dunne RF, Roeland EJ. The Interplay Among Pancreatic Cancer, Cachexia, Body Composition, and Diabetes. Hematol Oncol Clin North Am 2022; 36:897-910. [PMID: 36154783 DOI: 10.1016/j.hoc.2022.07.001] [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: 11/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with complex changes in body composition. Visceral obesity and type 2 diabetes mellitus are established risk factors for developing PDAC; however, clinical and metabolic features of PDAC commonly lead to cancer cachexia, a hypermetabolic syndrome characterized by weight loss secondary to muscle and adipose tissue wasting. Reduction in muscle mass in patients with PDAC is associated with poorer survival in patients undergoing surgical resection and increased chemotherapy toxicity. Although no standardized treatment exists, a multidisciplinary, tailored, symptom-based approach is recommended to improve outcomes and quality of life for patients with PDAC and cachexia.
Collapse
Affiliation(s)
- Richard F Dunne
- Department of Medicine, Wilmot Cancer Institute, University of Rochester Medical Center, 601 Elmwood Avenue, Box 704, Rochester, NY 14642, USA.
| | - Eric J Roeland
- Division of Hematology/Oncology, Oregon Health and Science University, Knight Cancer Institute, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
29
|
de Oliveira LC, Calixto-Lima L, Cunha GDC, Silva NFD, Souza-Silva RD, Fonseca TSM, Souza TD, Santos CDM, Santos DAD, Varea Maria Wiegert E. Effects of specialised nutritional interventions in patients with incurable cancer: a systematic review. BMJ Support Palliat Care 2022; 12:388-402. [DOI: 10.1136/spcare-2022-003893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/21/2022] [Indexed: 11/04/2022]
Abstract
ObjectiveTo evaluate current evidence of the effect of specialised nutritional interventions on nutritional status, survival, quality of life and measures of functionality in patients with incurable cancer.MethodsSystematic literature review according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines using PubMed/MEDLINE, EMBASE, Scopus, LILACS and Cochrane Library databases. Clinical studies that evaluated different specialised nutritional interventions, such as nutritional counselling, oral nutritional supplementation (ONS), enteral nutrition (EN) and parenteral nutrition (PN), were eligible. Only studies classified as being of high methodological quality (ie, low or moderate risk of bias) were included.ResultsA total of 22 studies reporting on 2448 patients were deemed eligible. Five types of specialised nutrition were observed: mixed (multimodal nature, ie, dietary counseling, ONS, physical activity and/or drugs) (n=12), ONS (n=5), PN (n=3), EN (n=1) and multidisciplinary team counselling (n=1). Benefits of any kind from the interventions were reported in 14 (63.6%) studies, mainly resulting from mixed intervention. Nutritional status improved in 12 (60.0%) of 20 studies and quality of life improved in eight (50.0%) of 16 studies. Few studies have evaluated the influence of nutritional interventions on survival and measure of functionality, and have not shown improvement in these outcomes.ConclusionDespite the limited evidence, specialised nutritional interventions can yield positive effects for patients with incurable cancer, mainly in their nutritional status and quality of life.
Collapse
|
30
|
Arslan B, Çolak T, Dağ A. Does Home Oral Nutritional Support Improve Nutritional Status and Quality of Life following Colorectal Cancer Surgery? Nutr Cancer 2022; 75:174-185. [PMID: 35852357 DOI: 10.1080/01635581.2022.2096911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Oral nutritional support (ONS) is a form of supportive intervention in patients' diet in response to insufficient oral intake, malabsorption, or functional insufficient food intake during the disease process. This study aimed to evaluate the nutritional status, morbidity, re-admissions, functional status, QoL of patients who had undergone elective colorectal cancer surgery and initiated home ONS. A total of 144 patients who had undergone colorectal cancer surgery and given home ONS were analyzed with regard to demographics, diagnosis, weight-BMI changes, re-admissions, morbidity, daily caloric and protein intake, functional status (Barthel index) and QoL (The Satisfaction with Life Scale-SWLS). The mean age was 65,6 ± 12,8 with a Female/Male ratio of 53/91. The mean BMI increased from 25.71 ± 3.81 to 28,35 ± 4,53 (p < 0.0001). Re-admission was significantly higher in patients who had received 600 kcal (55.8%) than those who received 900 kcal (40.2%) (p = 0.007). Furthermore, adaptation to chemotherapy (p = 0.02) and the Bartel index scores (p = 0.001) were significantly worse in patients who received 600 kcal compared to patients who received 900 kcal; however, the complication rate (p = 0.84), adaptation to radiotherapy (p = 0.68) and the QoL scores (p = 0.35) were not significantly different. Home ONS improved the BMI in all patients. In addition, ONS resulted in good outcomes with regard to adaptation to chemotherapy and the functional status in the treatments of patients with colorectal cancer.
Collapse
Affiliation(s)
- Bilal Arslan
- Department of Surgery, Division of Surgical Oncology, Mersin University Medical Faculty, Mersin, Turkey
| | - Tahsin Çolak
- Department of Surgery, Division of Surgical Oncology, Mersin University Medical Faculty, Mersin, Turkey
| | - Ahmet Dağ
- Department of Surgery, Division of Surgical Oncology, Mersin University Medical Faculty, Mersin, Turkey
| |
Collapse
|
31
|
Gupta P, Hodgman CF, Schadler KL, LaVoy EC. Effect of exercise on pancreatic cancer patients during treatment: a scoping review of the literature. Support Care Cancer 2022; 30:5669-5690. [PMID: 35190894 DOI: 10.1007/s00520-022-06925-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/16/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Exercise can lower the risk of developing pancreatic cancer and has the potential to improve physical fitness and quality of life in patients with the disease. Yet, the effects of exercise training during pancreatic cancer treatment remain poorly characterized. This hampers the development of evidence-based disease-specific exercise recommendations. PURPOSE The purpose of this review was to describe and interpret the effect of exercise on physiological, QoL, and cancer-specific outcomes reported in clinical trials among pancreatic cancer patients during treatment. METHODS We conducted a scoping review of the literature according to the framework proposed by Arksey and O'Malley. Articles published prior to December 2021 were retrieved from PubMed, EMBASE, and Scopus. We only included studies that prescribed structured cardiorespiratory and/or resistance exercise in pancreatic cancer patients undergoing treatment. RESULTS A total of 662 references were retrieved, of which 24 are included in the review. Twelve articles were randomized controlled trials and 12 were single-arm trials. Overlap in the trials from which data were reported occurred in 16 articles. Moderate intensity exercise was most commonly prescribed, reported feasible for most patients, with potential to enhance physical fitness and QoL. However, exercise adherence and beneficial effects may diminish with disease progression. Limited evidence suggests exercise may benefit cancer-specific outcomes. CONCLUSION The results of this review indicate that exercise is feasible during pancreatic cancer treatment. Exercise can also improve physical fitness and QoL. However, its beneficial effects may fall with advanced disease and more rigorous research is needed to develop precise exercise protocols for this population.
Collapse
Affiliation(s)
- Priti Gupta
- Department of Health and Human Performance, The University of Houston, 3875 Holman St., Rm 104 Garrison, Houston, TX, 77204-6015, USA
| | - Charles F Hodgman
- Department of Health and Human Performance, The University of Houston, 3875 Holman St., Rm 104 Garrison, Houston, TX, 77204-6015, USA
| | - Keri L Schadler
- Department of Pediatrics, MD Anderson Cancer Center, Houston, TX, USA
| | - Emily C LaVoy
- Department of Health and Human Performance, The University of Houston, 3875 Holman St., Rm 104 Garrison, Houston, TX, 77204-6015, USA.
| |
Collapse
|
32
|
Steindorf K, Clauss D, Rötzer I, Tjaden C, Hackert T, Wiskemann J. Nutrition Intake and Nutrition Status of Pancreatic Cancer Patients: Cross-Sectional and Longitudinal Analysis of a Randomized Controlled Exercise Intervention Study. Nutr Cancer 2022; 74:3492-3500. [PMID: 35608567 DOI: 10.1080/01635581.2022.2077382] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 12/16/2022]
Abstract
Pancreatic cancer patients often present with an inadequate nutritional intake. At the same time, there are no standardized recommendations for nutrition intake during and after cancer treatment. In a prospective analysis of a randomized controlled trial analyzing the effects of a 6-month resistance training in pancreatic cancer patients, we assessed the nutritional intake and the impact of a 6-month supervised resistance training or home-based resistance training vs. usual care control on the nutritional intake of the patients. Nutritional intake was assessed by 24-h recall before and after the 6-month resistance training period. At baseline low protein intake (<1 g/kg body weight) was found in 33.9% of the 59 patients and low energy intake (<25 kcal/kg body weight) was found in 39.0% of the patients. In all, 35.6% of the patients were classified with a risk of malnutrition (NRS ≥ 3). In the total of 46 patients who finished the 6-month intervention period, there was no difference in nutritional intake over time between resistance training and usual care control. In conclusion, it appears that the majority of our study population had an adequate protein and energy intake. A resistance training seems to have no influence on the nutritional intake of the patients.
Collapse
Affiliation(s)
- Karen Steindorf
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Dorothea Clauss
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
- Department of Molecular and Cellular Sport Medicine, German Sport University, Cologne, Germany
| | - Ingeborg Rötzer
- Division of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Christine Tjaden
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Joachim Wiskemann
- Division of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
33
|
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: 10] [Impact Index Per Article: 3.3] [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
|
34
|
Rich NE, Phen S, Desai N, Mittal S, Yopp AC, Yang JD, Marrero JA, Iyengar P, Infante RE, Singal AG. Cachexia is Prevalent in Patients With Hepatocellular Carcinoma and Associated With Worse Prognosis. Clin Gastroenterol Hepatol 2022; 20:e1157-e1169. [PMID: 34555519 PMCID: PMC8934317 DOI: 10.1016/j.cgh.2021.09.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/21/2021] [Accepted: 09/10/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Cancer cachexia is a wasting syndrome associated with functional impairment and reduced survival that impacts up to 50% of patients with gastrointestinal cancers. However, data are limited on the prevalence and clinical significance of cachexia in patients with hepatocellular carcinoma (HCC). METHODS We performed a retrospective cohort study of patients diagnosed with HCC at 2 United States health systems between 2008 and 2018. Patient weights were recorded 6 months prior to and at time of HCC diagnosis. Cachexia was defined as >5% weight loss (or >2% weight loss if body mass index <20 kg/m2), and precachexia was defined as 2% to 5% weight loss. We used multivariable logistic regression models to identify correlates of cachexia and multivariable Cox proportional hazard models to identify factors associated with overall survival. RESULTS Of 604 patients with HCC, 201 (33.3%) had precachexia and 143 (23.7%) had cachexia at diagnosis, including 19.0%, 23.5%, 34.7%, and 34.0% of patients with Barcelona Clinic Liver Cancer stages 0/A, B, C, and D, respectively. Patients with cachexia were less likely to receive HCC treatment (odds ratio, 0.38; 95% confidence interval, 0.21-0.71) and had worse survival than those with precachexia or stable weight (11.3 vs 20.4 vs 23.5 months, respectively; P < .001). Cachexia remained independently associated with worse survival (hazard ratio, 1.43; 95% confidence interval, 1.11-1.84) after adjusting for age, sex, race, ethnicity, Child Pugh class, alpha-fetoprotein, Barcelona Clinic Liver Cancer stage, and HCC treatment. CONCLUSIONS Nearly 1 in 4 patients with HCC present with cachexia, including many with compensated cirrhosis or early stage tumors. The presence of cancer-associated weight loss appears to be an early and independent predictor of worse outcomes in patients with HCC.
Collapse
Affiliation(s)
- Nicole E. Rich
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX
| | - Samuel Phen
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX
| | - Nirali Desai
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX
| | - Sukul Mittal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX
| | - Adam C. Yopp
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX,Department of Surgery, UT Southwestern, Dallas TX
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center and Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles CA
| | - Jorge A. Marrero
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX
| | - Puneeth Iyengar
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX,Department of Radiation Oncology, UT Southwestern, Dallas TX
| | - Rodney E. Infante
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX,Center for Human Nutrition, UT Southwestern, Dallas TX
| | - Amit G. Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern, Dallas TX,Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas TX
| |
Collapse
|
35
|
Emanuel A, Krampitz J, Rosenberger F, Kind S, Rötzer I. Nutritional Interventions in Pancreatic Cancer: A Systematic Review. Cancers (Basel) 2022; 14:2212. [PMID: 35565341 PMCID: PMC9101959 DOI: 10.3390/cancers14092212] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
(1) Background: Pancreatic cancer (PaCa) is directly related to malnutrition, cachexia and weight loss. Nutritional interventions (NI) are used in addition to standard therapy. The aim of this systematic review is to provide an overview of the types of NI and their effects. (2) Methods: We included RCTs with at least one intervention group receiving an NI and compared them with a control group with no NI, placebo or alternative treatment on cachexia, malnutrition or weight loss in patients with PaCa. Any available literature until 12 August 2021 was searched in the Pubmed and Cochrane databases. RCTs were sorted according to NI (parenteral nutrition, enteral nutrition, dietary supplements and mixed or special forms). (3) Results: Finally, 26 studies with a total of 2720 patients were included. The potential for bias was mostly moderate to high. Parenteral nutrition is associated with a higher incidence of complications. Enteral nutrition is associated with shorter length of stay in hospital, lower rate and development of complications, positive effects on cytokine rates and lower weight loss. Dietary supplements enriched with omega-3 fatty acids lead to higher body weight and lean body mass. (4) Conclusions: Enteral nutrition and dietary supplements with omega-3 fatty acids should be preferred in nutritional therapy of PaCa patients.
Collapse
Affiliation(s)
- Aline Emanuel
- Division of Nutrition Sciences, German University of Applied Sciences for Prevention and Health Management (DHfPG), 66123 Saarbruecken, Germany;
| | - Julia Krampitz
- Division of Psychology and Pedagogy, German University of Applied Sciences for Prevention and Health Management (DHfPG), 66123 Saarbruecken, Germany;
- Institute of Psychology, University of Innsbruck, 6020 Innsbruck, Austria
| | - Friederike Rosenberger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Division of Health Sciences, German University of Applied Sciences for Prevention and Health Management (DHfPG), 66123 Saarbruecken, Germany;
| | - Sabine Kind
- Division of Health Sciences, German University of Applied Sciences for Prevention and Health Management (DHfPG), 66123 Saarbruecken, Germany;
| | - Ingeborg Rötzer
- Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital, 69120 Heidelberg, Germany;
- Clinic for Oncology and Haemotology, Northwest Hospital, UCT-Cancer University Center, 60488 Frankfurt am Main, Germany
| |
Collapse
|
36
|
Sultana S, Bouyahya A, Rebezov M, Shariati MA, Balahbib A, Khouchlaa A, El Yaagoubi OM, Khaliq A, Omari NE, Bakrim S, Zengin G, Akram M, Khayrullin M, Bogonosova I, Mahmud S, Simal-Gandara J. Impacts of nutritive and bioactive compounds on cancer development and therapy. Crit Rev Food Sci Nutr 2022; 63:9187-9216. [PMID: 35416738 DOI: 10.1080/10408398.2022.2062699] [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: 11/03/2022]
Abstract
For persons who survive with progressive cancer, nutritional therapy and exercise may be significant factors to improve the health condition and life quality of cancer patients. Nutritional therapy and medications are essential to managing progressive cancer. Cancer survivors, as well as cancer patients, are mostly extremely encouraged to search for knowledge about the selection of diet, exercise, and dietary supplements to recover as well as maintain their treatment consequences, living quality, and survival of patients. A healthy diet plays an important role in cancer treatment. Different articles are studied to collect information and knowledge about the use of nutrients in cancer treatment as well as cancer prevention. The report deliberates nutrition and exercise strategies during the range of cancer care, emphasizing significant concerns during treatment of cancer and for patients of advanced cancer, but concentrating mostly on the requirements of the population of persons who are healthy or who have constant disease following their repossession from management. It also deliberates choice nutrition and exercise problems such as dietary supplements, food care, food selections, and weight; problems interrelated to designated cancer sites, and common questions about diet, and cancer survival. Decrease the side effects of medicines both during and after treatment.
Collapse
Affiliation(s)
- Sabira Sultana
- Department of Eastern Medicine, Government College University Faisalabad, Pakistan
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathology Biology, Faculty of Sciences, and Genomic Center of Human Pathology, Mohammed V University, Rabat, Morocco
| | - Maksim Rebezov
- V M Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Moscow, Russian Federation
- K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Mohammad Ali Shariati
- K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Abdelaali Balahbib
- Laboratory of Biodiversity, Ecology, and Genome, Faculty of Sciences, Mohammed V University, Rabat, Morocco
| | - Aya Khouchlaa
- Laboratory of Human Pathology Biology, Faculty of Sciences, and Genomic Center of Human Pathology, Mohammed V University, Rabat, Morocco
| | - Ouadie Mohamed El Yaagoubi
- Laboratory of Biochemistry, Environment and Agri-Food (URAC 36) - Faculty of Sciences and Techniques - Mohammedia, Hassan II University Casablanca - Morocco
| | - Adnan Khaliq
- Department of Food Science and Technology, Khwaja Fareed University of Engineering and Information Technology, Pakistan
| | - Nasreddine El Omari
- Laboratory of Histology, Embryology, and Cytogenetic, Faculty of Medicine and Pharmacy, Mohammed V University, Rabat, Morocco
| | - Saad Bakrim
- Molecular Engineering, Valorization and Environment Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, Morocco
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| | - Muhammad Akram
- Department of Eastern Medicine, Government College University Faisalabad, Pakistan
| | - Mars Khayrullin
- K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Irina Bogonosova
- K.G. Razumovsky Moscow State University of Technologies and Management (The First Cossack University), Moscow, Russian Federation
| | - Shafi Mahmud
- Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Jesus Simal-Gandara
- Department of Analytical Chemistry and Food Science, Faculty of Science, Universidade de Vigo, Nutrition and Bromatology Group, Ourense, Spain
| |
Collapse
|
37
|
Mäkitie AA, Alabi RO, Orell H, Youssef O, Almangush A, Homma A, Takes RP, López F, de Bree R, Rodrigo JP, Ferlito A. Managing Cachexia in Head and Neck Cancer: a Systematic Scoping Review. Adv Ther 2022; 39:1502-1523. [PMID: 35224702 PMCID: PMC8989808 DOI: 10.1007/s12325-022-02074-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/03/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Patients with head and neck cancer (HNC) are usually confronted with functional changes due to the malignancy itself or its treatment. These factors typically affect important structures involved in speech, breathing, chewing, swallowing, and saliva production. Consequently, the intake of food will be limited, which further contributes to loss of body weight and muscle mass, anorexia, malnutrition, fatigue, and anemia. This multifactorial condition can ultimately lead to cancer cachexia syndrome. This study aims to examine the treatment of cachexia in HNC patients. METHODS We systematically searched OvidMedline, PubMed, Scopus, and Web of Science for articles examining the treatment of cachexia in HNC. RESULTS A total of nine studies were found, and these suggested interventions including nutritional, pharmacologic, therapeutic exercise, and multimodal approaches. The nutritional intervention includes essential components such as dietary counseling, oral nutritional supplements, and medical nutritional support. Individualized nutritional interventions include oral, enteral (feeding tubes i.e., percutaneous endoscopic gastrostomy [PEG], nasogastric tube [NGT]) and parenteral nutrition. The pharmacologic interventions aim at increasing the appetite and weight of cachectic patients. Therapeutic exercise and increased physical activity can help to enhance the synthesis of muscle protein, reducing inflammation and the catabolic effects of cachexia syndrome. CONCLUSION Owing to the multifactorial nature of this syndrome, it is expected that the management approach should be multi-interventional. Early implementation of these interventions may help to improve survival and quality of health and life of cachectic HNC patients.
Collapse
Affiliation(s)
- Antti A Mäkitie
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 263, 00029 HUS, Helsinki, Finland.
- Faculty of Medicine, Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland.
- Division of Ear, Nose and Throat Diseases, Department of Clinical Sciences, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
| | - Rasheed Omobolaji Alabi
- Faculty of Medicine, Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
- Department of Industrial Digitalization, School of Technology and Innovations, University of Vaasa, Vaasa, Finland
| | - Helena Orell
- Clinical Nutrition Unit, Internal Medicine and Rehabilitation, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Omar Youssef
- Faculty of Medicine, Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki, Helsinki, Finland
| | - Alhadi Almangush
- Faculty of Medicine, Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
- Department of Pathology, University of Helsinki, Helsinki, Finland
- Institute of Biomedicine, Pathology, University of Turku, Turku, Finland
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Robert P Takes
- Department of Otolaryngology-Head and Neck Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Fernando López
- Department of Otolaryngology-Head and Neck Surgery, Hospital Universitario Central de Asturias, University of Oviedo, ISPA, IUOPA, CIBERONC, Oviedo, Spain
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Juan P Rodrigo
- Department of Otolaryngology-Head and Neck Surgery, Hospital Universitario Central de Asturias, University of Oviedo, ISPA, IUOPA, CIBERONC, Oviedo, Spain
| | - Alfio Ferlito
- Coordinator of the International Head and Neck Scientific Group, Padua, Italy
| |
Collapse
|
38
|
Aoyama T, Yoshikawa T, Ida S, Cho H, Sakamaki K, Ito Y, Fujitani K, Takiguchi N, Kawashima Y, Nishikawa K, Nunobe S, Hiki N. Effects of perioperative eicosapentaenoic acid‑enriched oral nutritional supplement on the long‑term oncological outcomes after total gastrectomy for gastric cancer. Oncol Lett 2022; 23:151. [PMID: 35836480 PMCID: PMC9258592 DOI: 10.3892/ol.2022.13272] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 12/24/2022] Open
Abstract
Basic and clinical reports have suggested that eicosapentaenoic acid (EPA) exhibits anti-tumor activity. The present study evaluated whether perioperative EPA could improve the survival of patients with localized gastric cancer as a key secondary endpoint of a randomized clinical study. The present study was designed as multicenter, open-label, superiority, randomized trial to confirm the preventive effect of EPA on body weight loss after total gastrectomy for gastric cancer. Eligible patients were randomized to either the standard-diet group (EPA-off group) or EPA-on group by a centralized dynamic method. An EPA-enriched supplement (ProSure®) was given to the EPA-on group in addition to their standard diet. This supplement included 600 kcal with 2.2 g/day of EPA. Among the 126 patients who were randomized, 123 patients (EPA-off group, n=60; EPA-on group, n=63) were examined in the survival analyses. All background factors were well balanced between the two groups. The 3-year and 5-year overall survival rates were 74.6 and 67.8%, respectively, in the EPA-off group, and 77.8 and 76.2% in the EPA-on group. There was no significant difference between the EPA-off and EPA-on groups (hazard ratio, 0.77; P=0.424). In the subgroup analysis, the hazard ratio was 0.39 in patients who received neoadjuvant chemotherapy and 0.57 in patients with nodal metastasis. In conclusion, a clear survival benefit of perioperative EPA was not observed in localized gastric cancer. The value of EPA should be further tested in a future study in patients with unfavorable advanced gastric cancer. Clinical trial number: UMIN000006380; date of registration, September 21, 2011.
Collapse
Affiliation(s)
- Toru Aoyama
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Takaki Yoshikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Satoshi Ida
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Haruhiko Cho
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Kentaro Sakamaki
- Department of Biostatistics and Epidemiology, Yokohama City University Medical Center, Yokohama, Kanagawa 232‑0024, Japan
| | - Yuichi Ito
- Department of Gastroenterological Surgery, Aichi Cancer Center, Nagoya, Aichi 464‑8681, Japan
| | - Kazumasa Fujitani
- Department of Surgery, Osaka General Medical Center, Osaka 558‑8558, Japan
| | - Nobuhiro Takiguchi
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chuo-ku, Chiba 260‑8781, Japan
| | - Yoshiyuki Kawashima
- Department of Gastroenterological Surgery, Saitama Cancer Center, Kitaadachi, Saitama 362‑0806, Japan
| | | | - Soya Nunobe
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| | - Naoki Hiki
- Department of Gastroenterological Surgery, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo 135‑0063, Japan
| |
Collapse
|
39
|
Counts BR, Halle JL, Carson JA. Early-Onset Physical Inactivity and Metabolic Dysfunction in Tumor-bearing Mice Is Associated with Accelerated Cachexia. Med Sci Sports Exerc 2022; 54:77-88. [PMID: 34431825 PMCID: PMC8678203 DOI: 10.1249/mss.0000000000002772] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
METHODS Male C57BL/6J mice (12 wk of age) were injected with 1 × 106 LLC cells or phosphate-buffered saline (PBS) subcutaneously in the right flank, and tissue was collected 26-28 d after cell injection. Tumor volume was measured every 5 d throughout the study to calculate the tumor growth rate. Fifteen days after tumor inoculation, a subset of PBS (n = 11) and LLC (n = 16) mice were individually housed in metabolic Comprehensive Laboratory Animal Monitoring System cages for 5 d. RESULTS LLC mice exhibited greater body weight loss (-5.1%), decreased muscle mass (-7%), decreased fat mass (-22%), and increased plasma interleukin-6 (212%) compared with PBS mice. Before the onset of cachexia, total cage activity was decreased in tumor-bearing mice. Cage activity was negatively associated with tumor mass and positively associated with hindlimb muscle mass. In addition, LLC mice had greater lipid oxidation than PBS mice. CONCLUSIONS LLC mice exhibit early-onset physical inactivity and altered systemic lipid oxidation, which are associated with the eventual development of cachexia.
Collapse
Affiliation(s)
- Brittany R Counts
- Integrative Muscle Biology Laboratory, Division of Rehabilitation Sciences, College of Health Professions, University of Tennessee Health Science Center, Memphis TN
| | | | | |
Collapse
|
40
|
Gaafer OU, Zimmers TA. Nutrition challenges of cancer cachexia. JPEN J Parenter Enteral Nutr 2021; 45:16-25. [PMID: 34897740 DOI: 10.1002/jpen.2287] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/13/2021] [Accepted: 10/19/2021] [Indexed: 12/11/2022]
Abstract
Cancer cachexia, or progressive weight loss, often despite adequate nutrition contributes greatly to cancer morbidity and mortality. Cachexia is metabolically distinct from starvation or protein malnutrition, although many patients with cancer and cachexia exhibit lowered appetite and food consumption. Tumors affect neural mechanisms that regulate appetite and energy expenditure, while promoting wasting of peripheral tissues via catabolism of cardiac and skeletal muscle, adipose, and bone. These multimodal actions of tumors on the host suggest a need for multimodal interventions. However, multiple recent consensus guidelines for management of cancer cachexia differ in treatment recommendations, highlighting the lack of effective, available therapies. Challenges to defining appropriate nutrition or other interventions for cancer cachexia include lack of consensus on definitions, low strength of evidence from clinical trials, and a scarcity of robust, rigorous, and mechanistic studies. However, efforts to diagnose, stage, and monitor cachexia are increasing along with clinical trial activity. Furthermore, preclinical models for cancer cachexia are growing more sophisticated, encompassing a greater number of tumor types in organ-appropriate contexts and for metastatic disease to model the clinical condition more accurately. It is expected that continued growth, investment, and coordination of research in this topic will ultimately yield robust biomarkers, clinically useful classification and staging algorithms, targetable pathways, pivotal clinical trials, and ultimately, cures. Here, we provide an overview of the clinical and scientific knowledge and its limitations around cancer cachexia.
Collapse
Affiliation(s)
- Omnia U Gaafer
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Teresa A Zimmers
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA.,Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, Indiana, USA.,Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA.,Richard L. Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA
| |
Collapse
|
41
|
Gresham G, Placencio‐Hickok VR, Lauzon M, Nguyen T, Kim H, Mehta S, Paski S, Pandol SJ, Osipov A, Gong J, Jamil LH, Nissen N, Lo SK, Hendifar AE. Feasibility and efficacy of enteral tube feeding on weight stability, lean body mass, and patient-reported outcomes in pancreatic cancer cachexia. J Cachexia Sarcopenia Muscle 2021; 12:1959-1968. [PMID: 34609081 PMCID: PMC8718084 DOI: 10.1002/jcsm.12799] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/06/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Advanced pancreatic ductal adenocarcinoma (PDAC) is characterized by progressive weight loss and nutritional deterioration. This wasting has been linked to poor survival outcomes, alterations in host defenses, decreased functional ability, and diminished health-related quality of life (HRQOL) in pancreatic cancer patients. There are currently no standardized approaches to the management of pancreatic cancer cachexia. This study explores the feasibility and efficacy of enteral tube feeding of a peptide-based formula to improve weight stability and patient-reported outcomes (PROs) in advanced PDAC patients with cachexia. METHODS This was a single-institution, single-arm prospective trial conducted between April 2015 and March 2019. Eligible patients were adults (>18 years) diagnosed with advanced or locally advanced PDAC and cachexia, defined as greater than 5% unexplained weight loss within 6 months from screening. The study intervention included three 28 day cycles of a semi-elemental peptide-based formula, administered through a jejunal or gastrojejunal feeding tube. The primary outcome was weight stability at 3 months (Cycle 3), defined as weight change less than 0.1 kg/baseline BMI unit from baseline. Secondary outcomes included changes in lean body mass, appendicular lean mass, bone mineral density, fat mass, and percent body fat, as measured with a DEXA scan, HRQOL (EORTC QLQC30) and NIH PROMIS PROs assessed at each cycle. Daily activity (steps, distance, active minutes, heart rate, and sleep) were remotely monitored using a wearable activity monitor (Fitbit) over the 3 month study period. RESULTS Thirty-six patients were screened for eligibility, 31 patients consented onto study and underwent jejunal tube placement, and 16 patients completed treatment: mean age 67 years (SD 9.3), 43.8% male. Among evaluable patients (n = 16), weight stability was achieved in 10 patients (62.5%), thus completing the trial early. Increases in lean body mass (1273.1, SD: 4078, P = 0.01) and appendicular lean mass (0.45, SD: 0.6, P = 0.02) were observed. Statistically significant improvements at Cycle 3 from baseline were also observed for QLQC30 role function [mean difference (MD): 20.1, P = 0.03], appetite (MD: 27.4, P = 0.02), and global health scores (MD: 13.3, P = 0.05) as well as for NIH PROMIS t-scores for depression (MD: -10.4, P = 0.006) and pain interference (MD: -7.5, P = 0.05). Objectively monitored (Fitbit) activity levels increased, although statistical significance was not reached. CONCLUSIONS Our findings suggest that enteral nutrition support may improve weight stability, lean body mass, appendicular lean mass and PROs in PDAC patients with cachexia who completed treatment, representing a subsample of the study population. The feasibility and role of enteral feeding in routine care remain unclear, and larger and randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Gillian Gresham
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | | | - Marie Lauzon
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Tyra Nguyen
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Haesoo Kim
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Sejal Mehta
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Shirley Paski
- Department of Medicine, Karsh Division of Gastroenterology and HepatologyCedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Stephen J. Pandol
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Arsen Osipov
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Jun Gong
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Laith H. Jamil
- Section of Gastroenterology and HepatologyBeaumont HealthRoyal OakMIUSA
- Oakland University William Beaumont School of MedicineRochesterMIUSA
| | - Nicholas Nissen
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Simon K. Lo
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| | - Andrew E. Hendifar
- Department of MedicineSamuel Oschin Comprehensive Cancer Institute, Cedars‐Sinai Medical CenterLos AngelesCAUSA
| |
Collapse
|
42
|
Bird JK, Troesch B, Warnke I, Calder PC. The effect of long chain omega-3 polyunsaturated fatty acids on muscle mass and function in sarcopenia: A scoping systematic review and meta-analysis. Clin Nutr ESPEN 2021; 46:73-86. [PMID: 34857251 DOI: 10.1016/j.clnesp.2021.10.011] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Sarcopenia is characterized by the progressive loss of skeletal muscle mass and function, which reduces mobility and quality of life. Risk factors for sarcopenia include advanced age, physical inactivity, obesity, and chronic diseases such as cancer or rheumatoid arthritis. Omega-3 long chain polyunsaturated fatty acids (LC PUFAs) might be associated with a reduction in risk of sarcopenia due to their anti-inflammatory effects. METHODS We conducted a systematic review and meta-analysis to quantify the effects of omega-3 LC PUFAs on muscle mass, volume and function parameters. The National Library of Medicine's MEDLINE/PubMed database was searched on 9th October 2020 for randomized controlled trials that used omega-3 LC PUFAs as an intervention with muscle-related endpoints. A snowballing search to identify additional studies was completed on 23rd April 2021. The meta-analysis was conducted using meta-essentials worksheet 3. Bias was assessed using the Jadad scale. RESULTS 123 studies were identified with the systematic searches. Most studies were performed in disease populations, such as cancer or chronic obstructive pulmonary disease (COPD), or in healthy individuals after a fatiguing exercise bout. The endpoints lean body mass, skeletal muscle mass, mid-arm muscle circumference, handgrip strength, quadriceps maximal voluntary capacity (MVC), and 1-repetition maximum chest press were selected for meta-analysis based on the number of available studies; thus 66 studies were included in the quantitative synthesis. Using a random effects model and 2-tailed p-value, there was a significant relationship in favor of omega-3 LC PUFA supplementation for lean body mass (effect size 0.27, 95%CI 0.04 to 0.51), skeletal muscle mass (effect size 0.31, 95%CI 0.01 to 0.60) and quadriceps MVC (effect size 0.47, 95%CI 0.02 to 0.93). CONCLUSION The results indicate that there is a positive effect of omega-3 LC PUFA supplementation on overall body muscle mass and strength. Small study size and heterogeneity limit the applicability of these findings for sarcopenia prevention. Larger trials in populations at risk of sarcopenia would strengthen the evidence base.
Collapse
Affiliation(s)
- Julia K Bird
- Bird Scientific Writing, Wassenaar, 2242, the Netherlands.
| | - Barbara Troesch
- Nutrition Science and Advocacy, DSM Nutritional Products, 4303, Kaiseraugst, Switzerland.
| | - Ines Warnke
- R&D Human Nutrition and Health, DSM Nutritional Products, 4303, Kaiseraugst, Switzerland.
| | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, United Kingdom; NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, SO16 6YD, United Kingdom.
| |
Collapse
|
43
|
Wang Y, Liu R, Chang M, Wei W, Guo Y, Jin Q, Wang X. Does omega-3 PUFA-enriched oral nutritional intervention benefit cancer patients receiving chemo (radio) therapy? A systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr 2021:1-16. [PMID: 34606391 DOI: 10.1080/10408398.2021.1984199] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Although data indicate omega-3 polyunsaturated fatty acids are beneficial nutrients in cancer therapy, the evidences for efficacy of nutritional interventions during chemo (radio) therapy are still limited. The leading goal of the present meta-analysis was to summarize randomized controlled trials involving the administration of ω-3 PUFA-enriched oral nutritional supplements during chemo (radio) therapy, and evaluate the effects on nutritional status and clinical outcomes in patients. We systematically searched PubMed, Embase, Web of Science, Cochrane databases to identify interventions assessing body weight, BMI, immune and inflammatory indicators, plasma omega-3 fatty acids and adverse events, with subgroup analyses for region, types of ω-3 fatty acids, dose, duration and dosage form. In total, 22 studies including 1155 participants met the inclusion criteria. Meta-analysis showed a significant increase in body weight (BW) (WMD = 0.59 kg, 95% CI: 0.06, 1.13, P = 0.03), body mass index (BMI) (WMD = 0.43 kg/m2, 95% CI: 0.07, 0.79, P = 0.02), and plasma total ω-3 fatty acids (SMD = 2.52, 95% CI: 1.27, 3.78, P<0.0001), and a significant reduction in plasma levels of C-reactive protein (CRP) (SMD= -0.53, 95% CI: -0.80, -0.25, P = 0.0001), tumor necrosis factor-α (TNF-α) (WMD = -0.40 pg/mL, 95% CI: -0.80, -0.01, P = 0.05), interleukin 6 (IL-6) (WMD = -1.25 pg/mL, 95% CI: -2.41, -0.10, P = 0.03) and the incidence of adverse events (RR= 0.72, 95% CI: 0.54, 0.95, P = 0.02). However, plasma albumin levels (WMD = 0.02 mg/dL, 95% CI: -0.13, 0.18, P = 0.75) was remained unaffected. Overall, our meta-analysis provides evidences that the consumption of ω-3 PUFA-enriched oral nutritional supplements exert beneficial effects on nutritional status and clinical outcomes in patients undergoing chemo (radio) therapy.
Collapse
Affiliation(s)
- Yandan Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ruijie Liu
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ming Chang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Wei
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Yiwen Guo
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qingzhe Jin
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xingguo Wang
- National Engineering Research Center for Functional Food, Collaborative Innovation Center of Food Safety and Quality Control in Jiangsu Province, State Key Lab of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
44
|
Purcell SA, Marker RJ, Cornier MA, Melanson EL. Dietary Intake and Energy Expenditure in Breast Cancer Survivors: A Review. Nutrients 2021; 13:nu13103394. [PMID: 34684403 PMCID: PMC8540510 DOI: 10.3390/nu13103394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Many breast cancer survivors (BCS) gain fat mass and lose fat-free mass during treatment (chemotherapy, radiation, surgery) and estrogen suppression therapy, which increases the risk of developing comorbidities. Whether these body composition alterations are a result of changes in dietary intake, energy expenditure, or both is unclear. Thus, we reviewed studies that have measured components of energy balance in BCS who have completed treatment. Longitudinal studies suggest that BCS reduce self-reported energy intake and increase fruit and vegetable consumption. Although some evidence suggests that resting metabolic rate is higher in BCS than in age-matched controls, no study has measured total daily energy expenditure (TDEE) in this population. Whether physical activity levels are altered in BCS is unclear, but evidence suggests that light-intensity physical activity is lower in BCS compared to age-matched controls. We also discuss the mechanisms through which estrogen suppression may impact energy balance and develop a theoretical framework of dietary intake and TDEE interactions in BCS. Preclinical and human experimental studies indicate that estrogen suppression likely elicits increased energy intake and decreased TDEE, although this has not been systematically investigated in BCS specifically. Estrogen suppression may modulate energy balance via alterations in appetite, fat-free mass, resting metabolic rate, and physical activity. There are several potential areas for future mechanistic energetic research in BCS (e.g., characterizing predictors of intervention response, appetite, dynamic changes in energy balance, and differences in cancer sub-types) that would ultimately support the development of more targeted and personalized behavioral interventions.
Collapse
Affiliation(s)
- Sarah A. Purcell
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.-A.C.); (E.L.M.)
- Anschutz Health and Wellness Center, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
- Correspondence:
| | - Ryan J. Marker
- Anschutz Health and Wellness Center, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
- Department of Physical Medicine and Rehabilitation, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| | - Marc-Andre Cornier
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.-A.C.); (E.L.M.)
- Anschutz Health and Wellness Center, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA;
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Edward L. Melanson
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA; (M.-A.C.); (E.L.M.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Geriatric Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA
| |
Collapse
|
45
|
Turcott JG, Zatarain-Barrón ZL, Cárdenas Fernández D, Castañares Bolaños DT, Arrieta O. Appetite stimulants for patients with cancer: current evidence for clinical practice. Nutr Rev 2021; 80:857-873. [PMID: 34389868 DOI: 10.1093/nutrit/nuab045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The incidence of neoplastic diseases has increased worldwide, with an estimated global burden of 19.3 million incident cases and 10 million deaths in 2020-a considerable increase compared with 9.6 million deaths in 2018. One of the most prevalent problems faced by patients with cancer and their physicians is malnutrition. It is estimated that patients with cancer have important nutritional alterations in 25% to 70% of cases, which directly affects many spheres of patient care and well-being, including quality of life, treatment toxicity, and survival outcomes. Despite the overwhelming need to address this pressing issue, current evidence in terms of pharmacologic interventions for cancer-related anorexia remains inconclusive, and there is no current standard of care for patients with cancer-related anorexia. Nonetheless, international guidelines recommend promoting anabolism through nutritional, physical, and pharmacologic therapies. In this review, the available information is summarized regarding pharmacologic therapies to treat cancer-related anorexia and findings are highlighted from a clinical stance.
Collapse
Affiliation(s)
- Jenny G Turcott
- Thoracic Oncology Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | | | | | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
46
|
Changes in Physical Function and Effects on QOL in Patients after Pancreatic Cancer Surgery. Healthcare (Basel) 2021; 9:healthcare9070882. [PMID: 34356260 PMCID: PMC8304148 DOI: 10.3390/healthcare9070882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023] Open
Abstract
This study examined the changes in physical function and quality of life (QOL) of postoperative patients with pancreatic cancer for 3 months after surgery and examined the factors affecting the QOL at the 3 months after surgery. Methods: This study comprised 32 pancreatic cancer patients who underwent surgery at our hospital. Among these patients, 20 patients for whom data was measured before surgery to 3 months after surgery were selected for statistical analyses: 8 males and 12 females, 69.8 ± 7.4 years. The preoperative and postoperative rehabilitation was given to patients under the guidance of a physiotherapist. Nutritional status, body composition, physical function, gait assessments, and QOL were investigated. Results: Body weight, body fat mass, body fat percentage, body mass index (BMI), and muscle mass significantly decreased 3 months after surgery compared with their respective preoperative values. The mean grip strength at the time of 3 months after the surgery had decreased significantly from 27.3 kg to 24.5 kg. The mean skeletal muscle mass index (SMI) had decreased significantly from 6.3 kg before surgery to 5.9 kg after the surgery. The QOL scores for global health status, physical, and role showed significant decreases 2 weeks after surgery compared with the respective preoperative scores. Significant improvements in these scores were observed 3 months after surgery compared with the respective scores 2 weeks after surgery. Physical function assessments after surgery were associated with QOL 3 months after surgery. Conclusion: Recovery of patients after pancreatic cancer surgery in body weight, BMI, body fat percentage, body fat percentage, muscle mass, SMI, and grip strength was not sufficient at the time of 3 months after surgery. It has been observed that physical function of patients has affected the improvement of QOL.
Collapse
|
47
|
Fix DK, Counts BR, Smuder AJ, Sarzynski MA, Koh H, Carson JA. Wheel running improves fasting-induced AMPK signaling in skeletal muscle from tumor-bearing mice. Physiol Rep 2021; 9:e14924. [PMID: 34270178 PMCID: PMC8284248 DOI: 10.14814/phy2.14924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
Disruptions to muscle protein turnover and metabolic regulation contribute to muscle wasting during the progression of cancer cachexia. The initiation of cachexia is also associated with decreased physical activity. While chronic muscle AMPK activation occurs during cachexia progression in ApcMin/+ (MIN) mice, a preclinical cachexia model, the understanding of muscle AMPK's role during cachexia initiation is incomplete. Therefore, we examined if voluntary wheel exercise could improve skeletal muscle AMPK signaling in pre-cachectic MIN mice. Next, we examined muscle AMPK's role in aberrant catabolic signaling in response to a 12-h fast in mice initiating cachexia. Male C57BL/6 (B6: N = 26) and MIN (N = 29) mice were subjected to ad libitum feeding, 12-h fast, or 4 wks. of wheel access and then a 12-h fast during the initiation of cachexia. Male tamoxifen-inducible skeletal muscle AMPKα1 α2 (KO) knockout mice crossed with ApcMin/+ and floxed controls were examined (WT: N = 8, KO: N = 8, MIN: N = 10, MIN KO: N = 6). Male mice underwent a 12-h fast and the gastrocnemius muscle was analyzed. MIN gastrocnemius mass was reduced compared to B6 mice. A 12-h fast induced MIN muscle AMPKT172 , FOXOS413 , and ULK-1S555 phosphorylation compared to B6. Wheel running attenuated these inductions. A 12-h fast induced MIN muscle MuRF-1 protein expression compared to B6 and was suppressed by wheel running. Additionally, fasting induced muscle autophagy signaling and disrupted mitochondrial quality protein expression in the MIN, which was prevented in the MIN KO. We provide evidence that increased skeletal muscle AMPK sensitivity to a 12-h fast is an adverse event in pre-cachectic MIN mice, and exercise can improve this regulation.
Collapse
Affiliation(s)
- Dennis K. Fix
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - Brittany R. Counts
- Integrative Muscle Biology LaboratoryDivision of Rehabilitation SciencesCollege of Health ProfessionsUniversity of Tennessee Health Science CenterMemphisTNUSA
| | - Ashley J. Smuder
- Department of Applied Physiology & KinesiologyCollege of Health & Human PerformanceUniversity of FloridaGainesvilleFLUSA
| | - Mark A. Sarzynski
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - Ho‐Jin Koh
- Department of Exercise ScienceArnold School of Public HealthUniversity of South CarolinaColumbiaSCUSA
| | - James A. Carson
- Integrative Muscle Biology LaboratoryDivision of Rehabilitation SciencesCollege of Health ProfessionsUniversity of Tennessee Health Science CenterMemphisTNUSA
| |
Collapse
|
48
|
Hasegawa Y, Ijichi H, Saito K, Ishigaki K, Takami M, Sekine R, Usami S, Nakai Y, Koike K, Kubota N. Protein intake after the initiation of chemotherapy is an independent prognostic factor for overall survival in patients with unresectable pancreatic cancer: A prospective cohort study. Clin Nutr 2021; 40:4792-4798. [PMID: 34265503 DOI: 10.1016/j.clnu.2021.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/26/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS This study was conducted to investigate the nutritional status and longitudinal dietary intake during the course of chemotherapy, and their relationships with the survival in patients with unresectable pancreatic cancer. METHODS A prospective cohort study was conducted in 38 patients with unresectable pancreatic cancer receiving chemotherapy between January 2018 and November 2019. Subjective global assessment was used to assess the nutritional status, and the dietary intake was assessed monthly, for up to 12 months, using a brief self-administered diet history questionnaire. The primary outcome was overall survival, and the secondary outcome was progression-free survival. Cox regression analysis was performed to identify independent prognostic factors. RESULTS Moderate or severe malnutrition was found in 34.2% of the participants. Daily protein intake was significantly higher in the survivor group than in the deceased group at one month after the initiation of chemotherapy (1.4 ± 0.7 g/kg/day vs. 0.9 ± 0.5 g/kg/day, p = 0.019), while the baseline nutritional intakes were similar between the two groups. Univariate analysis identified weight loss >3.5%, energy intake <25 kcal/kg/day, protein intake <1.1 g/kg/day, and malnutrition as possible poor prognostic factors. Multivariate analysis identified protein intake <1.1 g/kg/day (hazard ratio [HR]: 9.03, 95%CI: 1.45-56.32, p = 0.018) as an independent poor prognostic factor. CONCLUSIONS Insufficient protein intake was identified as an independent poor prognostic factor in patients with unresectable pancreatic cancer receiving chemotherapy. Improving the dietary protein intake could be a useful therapeutic approach in patients with advanced pancreatic cancer receiving chemotherapy.
Collapse
Affiliation(s)
- Yoko Hasegawa
- Clinical Nutrition Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Hideaki Ijichi
- Clinical Nutrition Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Kei Saito
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazunaga Ishigaki
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Maki Takami
- Clinical Nutrition Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Rie Sekine
- Clinical Nutrition Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Satoshi Usami
- Graduate School of Education, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yousuke Nakai
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Endoscopy and Endoscopic Surgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhiko Koike
- Department of Gastroenterology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Naoto Kubota
- Clinical Nutrition Center, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan; Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
49
|
Comparison of total and activity energy expenditure estimates from physical activity questionnaires and doubly labelled water: a systematic review and meta-analysis. Br J Nutr 2021; 125:983-997. [PMID: 32718378 DOI: 10.1017/s0007114520003049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Physical activity questionnaires (PAQ) could be suitable tools in free-living people for measures of physical activity, total and activity energy expenditure (TEE and AEE). This meta-analysis was performed to determine valid PAQ for estimating TEE and AEE using doubly labelled water (DLW). We identified data from relevant studies by searching Google Scholar, PubMed and Scopus databases. This revealed thirty-eight studies that had validated PAQ with DLW and reported the mean differences between PAQ and DLW measures of TEE (TEEDLW - TEEPAQ) and AEE (AEEDLW - AEEPAQ). We assessed seventy-eight PAQ consisting of fifty-nine PAQ that assessed TEE and thirty-five PAQ that examined AEE. There was no significant difference between TEEPAQ and TEEDLW with a weighted mean difference of -243·3 and a range of -841·4 to 354·6 kJ/d, and a significant weighted mean difference of AEEDLW - AEE PAQ 414·6 and a range of 78·7-750·5. To determine whether any PAQ was a valid tool for estimating TEE and AEE, we carried out a subgroup analysis by type of PAQ. Only Active-Q, administered in two seasons, and 3-d PA diaries were correlated with TEE by DLW at the population level; however, these two PAQ did not demonstrate an acceptable limit of agreement at individual level. For AEE, no PAQ was correlated with DLW either at the population or at the individual levels. Active-Q and 3-d PA diaries were identified as the only valid PAQ for TEE estimation. Further well-designed studies are needed to verify this result and identify additional valid PAQ.
Collapse
|
50
|
Amarasinghe KC, Lopes J, Beraldo J, Kiss N, Bucknell N, Everitt S, Jackson P, Litchfield C, Denehy L, Blyth BJ, Siva S, MacManus M, Ball D, Li J, Hardcastle N. A Deep Learning Model to Automate Skeletal Muscle Area Measurement on Computed Tomography Images. Front Oncol 2021; 11:580806. [PMID: 34026597 PMCID: PMC8138051 DOI: 10.3389/fonc.2021.580806] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 03/22/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Muscle wasting (Sarcopenia) is associated with poor outcomes in cancer patients. Early identification of sarcopenia can facilitate nutritional and exercise intervention. Cross-sectional skeletal muscle (SM) area at the third lumbar vertebra (L3) slice of a computed tomography (CT) image is increasingly used to assess body composition and calculate SM index (SMI), a validated surrogate marker for sarcopenia in cancer. Manual segmentation of SM requires multiple steps, which limits use in routine clinical practice. This project aims to develop an automatic method to segment L3 muscle in CT scans. METHODS Attenuation correction CTs from full body PET-CT scans from patients enrolled in two prospective trials were used. The training set consisted of 66 non-small cell lung cancer (NSCLC) patients who underwent curative intent radiotherapy. An additional 42 NSCLC patients prescribed curative intent chemo-radiotherapy from a second trial were used for testing. Each patient had multiple CT scans taken at different time points prior to and post- treatment (147 CTs in the training and validation set and 116 CTs in the independent testing set). Skeletal muscle at L3 vertebra was manually segmented by two observers, according to the Alberta protocol to serve as ground truth labels. This included 40 images segmented by both observers to measure inter-observer variation. An ensemble of 2.5D fully convolutional neural networks (U-Nets) was used to perform the segmentation. The final layer of U-Net produced the binary classification of the pixels into muscle and non-muscle area. The model performance was calculated using Dice score and absolute percentage error (APE) in skeletal muscle area between manual and automated contours. RESULTS We trained five 2.5D U-Nets using 5-fold cross validation and used them to predict the contours in the testing set. The model achieved a mean Dice score of 0.92 and an APE of 3.1% on the independent testing set. This was similar to inter-observer variation of 0.96 and 2.9% for mean Dice and APE respectively. We further quantified the performance of sarcopenia classification using computer generated skeletal muscle area. To meet a clinical diagnosis of sarcopenia based on Alberta protocol the model achieved a sensitivity of 84% and a specificity of 95%. CONCLUSIONS This work demonstrates an automated method for accurate and reproducible segmentation of skeletal muscle area at L3. This is an efficient tool for large scale or routine computation of skeletal muscle area in cancer patients which may have applications on low quality CTs acquired as part of PET/CT studies for staging and surveillance of patients with cancer.
Collapse
Affiliation(s)
- Kaushalya C. Amarasinghe
- Bioinformatics Core Facility, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Jamie Lopes
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Julian Beraldo
- Radiation Therapy, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Nicole Kiss
- Institute for Physical Activity and Nutrition (IPAN), Deakin University, Geelong, VIC, Australia
- Allied Health Department, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Nicholas Bucknell
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Sarah Everitt
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Radiation Therapy, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Price Jackson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Cassandra Litchfield
- Bioinformatics Core Facility, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Linda Denehy
- Allied Health Department, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Melbourne School of Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin J. Blyth
- Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shankar Siva
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael MacManus
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Ball
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
- Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Jason Li
- Bioinformatics Core Facility, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Nicholas Hardcastle
- Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|