1
|
Mannino A, Lasry C, Kuypers J, Haines TP, Croagh D, Hanna L, Furness K. The effects of enteral tube feeding on nutrition, survival, and quality of life outcomes in advanced upper gastrointestinal cancers: a systematic literature review. Support Care Cancer 2025; 33:223. [PMID: 40009216 PMCID: PMC11865217 DOI: 10.1007/s00520-025-09263-6] [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: 07/26/2024] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
PURPOSE Despite guidelines, enteral tube feeding is not routinely provided to advanced upper gastrointestinal (UGI) cancer patients who cannot consume adequate nutrition and who have an expected survival of at least 3 months. This review examined its effect on nutrition status, survival, and quality of life (QOL) in these patients. METHODS Five databases (CINAHL, Cochrane, Embase, Ovid, Web of Science) were searched for original research on nutrition, survival, and/or QOL outcomes in adults with inoperable UGI cancers receiving enteral tube feeding. Quality was assessed using the Academy of Nutrition and Dietetics Quality Criteria Checklist: Primary Research, and a narrative synthesis was conducted. RESULTS Five studies were eligible for inclusion, most participants were male (n = 205), with low sample sizes across all studies (n = 16-131). Enteral tube feeding resulted in a similar proportion of participants with weight loss above or below 5% (baseline to 12 weeks) compared to a control group [p > 0.05] (1 study), and a significant increase in mean lean body mass [+1.3 (± 4.0) kg, p = 0.01] (1 study). There was variability in survival outcomes, statistical modelling, and comparators in five studies, with subsequently contradictory results. Only one study reported on QOL. Study quality was assessed as neutral (4 studies) or negative (1 study), reflecting methodological/analytical issues across the studies. CONCLUSIONS This systematic literature review highlights a significant knowledge gap, with no high-quality randomised controlled trial-based evidence available on enteral nutrition efficacy, limiting its use in dietetic practice in this sub-population. Despite treatment developments prolonging survival, research investigating feeding and its impact on QOL remains inadequate. Further research is needed to promote change and influence practice, policy, and guidelines, alongside high-quality intervention studies with defined nutrition outcomes, regimens, and robust statistical analyses to determine the benefits of enteral tube feeding in this vulnerable population.
Collapse
Affiliation(s)
- Adriana Mannino
- Department of Food, Nutrition and Dietetics, School Allied Health, Human Services and Sport, La Trobe University, Plenty Rd & Kingsbury Dr, Bundoora, Melbourne, Victoria, 3086, Australia
| | - Caroline Lasry
- Department of Food, Nutrition and Dietetics, School Allied Health, Human Services and Sport, La Trobe University, Plenty Rd & Kingsbury Dr, Bundoora, Melbourne, Victoria, 3086, Australia
| | - Julia Kuypers
- Department of Food, Nutrition and Dietetics, School Allied Health, Human Services and Sport, La Trobe University, Plenty Rd & Kingsbury Dr, Bundoora, Melbourne, Victoria, 3086, Australia
- Nutrition and Dietetics, St Vincents Hospital, Fitzroy, Melbourne, Victoria, 3065, Australia
| | - Terry P Haines
- School of Primary and Allied Health Care &, Faculty of Medicine, Nursing and Health Sciences, National Centre for Healthy Ageing, Monash University, Moorooduc Highway, Frankston, Victoria, 3199, Australia
| | - Daniel Croagh
- Department of Surgery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3168, Australia
| | - Lauren Hanna
- Department of Nutrition, Dietetics and Food, Monash University, Clayton, Victoria, 3168, Australia
| | - Kate Furness
- Department of Food, Nutrition and Dietetics, School Allied Health, Human Services and Sport, La Trobe University, Plenty Rd & Kingsbury Dr, Bundoora, Melbourne, Victoria, 3086, Australia.
| |
Collapse
|
2
|
Nguyen G, Furness K, Croagh D, Haines T, Hanna L. Supplemental tube feeding: qualitative study of patient perspectives in advanced pancreatic cancer. BMJ Support Palliat Care 2024; 14:e3002-e3010. [PMID: 38580394 PMCID: PMC11671877 DOI: 10.1136/spcare-2024-004836] [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: 02/18/2024] [Accepted: 03/12/2024] [Indexed: 04/07/2024]
Abstract
OBJECTIVES Malnutrition is associated with poor quality of life and survival outcomes for patients with cancer, but is challenging to prevent or treat in pancreatic cancer due to the multifactorial drivers of nutritional decline. A novel application of percutaneous endoscopic gastrostomy with a jejunal extension tube to deliver supplementary nutrition may improve outcomes, and will be tested in a randomised controlled trial. This study explored the perspectives of people living with pancreatic cancer regarding the acceptability of this proposed intensive nutrition intervention, to elucidate appropriateness and anticipated barriers, and facilitate informed design of the planned trial. METHODS Participants were patients with pancreatic cancer previously enrolled in a Pancreaticobiliary Cancer Biobank. Qualitative semi-structured interviews were conducted by telephone and transcribed verbatim for deductive thematic analysis. The Framework Model was used, with the Theoretical Framework of Acceptability as the analytical framework. RESULTS 10 participants were recruited. Four overarching themes were developed from interviews: (1) deterioration in physical and mental well-being are consequences of debilitating nutrition impact symptoms; (2) willingness to participate depends on an individual threshold for nutritional deterioration; (3) predicted perceived effectiveness outweighed anticipated burdens and (4) adequate dietetic support is needed for maintaining a percutaneous endoscopic gastrostomy with jejunal extension feeding tube at home with confidence. CONCLUSIONS Most participants believed that the intervention would benefit people with advanced pancreatic cancer to maintain their nutrition throughout chemotherapy. Regular and ad hoc support was considered essential, and the degree of individual nutritional deterioration was identified as an important indicator for trial participation.
Collapse
Affiliation(s)
- Grace Nguyen
- Melbourne Medical School, The University of Melbourne, Parkville, Victoria, Australia
| | - Kate Furness
- Department of Sport, Exercise and Nutrition Sciences, La Trobe University, Bundoora, Victoria, Australia
| | - Daniel Croagh
- Upper Gastrointestinal and Hepatobiliary Surgery, Monash Health, Clayton, Victoria, Australia
- Department of Surgery, Monash University, Clayton, Victoria, Australia
| | - Terry Haines
- School of Primary and Allied Health Care, Monash University, Frankston, Victoria, Australia
| | - Lauren Hanna
- Department of Nutrition, Dietetics and Food, Monash University, Notting Hill, Victoria, Australia
| |
Collapse
|
3
|
Bozzetti F. Evolving concepts on perioperative nutrition of sarcopenic cancer patients. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:106748. [PMID: 36376142 DOI: 10.1016/j.ejso.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022]
Abstract
The recent recognition of the association of sarcopenia with an increased risk of complications after a surgical procedure calls for rethinking the proper approach of the perioperative care in cancer patients. Sarcopenia is broadly considered in literature according to three different definitions: loss of muscle mass, loss of muscle mass plus reduced muscle function and myosteatosis. The aim of this short review on this issue is to define the excess of risk by type of primary and of surgical procedure, depending on the definition of sarcopenia, to speculate on this association (casual versus causal) and to examine the current therapeutical approaches. The analysis of the data shows that sarcopenia, defined as loss of muscle mass plus reduced muscle function, has the higher predictive power for the occurrence of postoperative complications than the two other definitions, and any definition of sarcopenia works better than the usual indexes or scores of surgical risk. Our analysis supports the concept that: a) sarcopenia is frequently associated with inflammation, but inflammation cannot be considered the only or the absolute cause for sarcopenia, b) sarcopenia is not a simple marker of risk but can have a direct role in the increase of risk. Data on perioperative care of sarcopenic cancer patients are scanty but a correct approach cannot rely on nutritional support alone but on a combined approach of optimized nutrition and exercise, hopefully associated with an anti-inflammatory treatment. This strategy should be applied proactively in keeping with the recent recommendations of the American Society of Clinical Oncology for the medical treatment of advanced cancer patients even if a clear demonstration of effectiveness is still lacking.
Collapse
Affiliation(s)
- Federico Bozzetti
- University of Milan, Faculty of Medicine, via Festa del Perdono, 20100, Milano, Italy.
| |
Collapse
|
4
|
Wu HF, Yan JP, Wu Q, Yu Z, Xu HX, Song CH, Guo ZQ, Li W, Xiang YJ, Xu Z, Luo J, Cheng SQ, Zhang FM, Shi HP, Zhuang CL. Discovery of distinct cancer cachexia phenotypes using an unsupervised machine-learning algorithm. Nutrition 2024; 119:112317. [PMID: 38154396 DOI: 10.1016/j.nut.2023.112317] [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: 08/01/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/30/2023]
Abstract
OBJECTIVES Cancer cachexia is a debilitating condition with widespread negative effects. The heterogeneity of clinical features within patients with cancer cachexia is unclear. The identification and prognostic analysis of diverse phenotypes of cancer cachexia may help develop individualized interventions to improve outcomes for vulnerable populations. The aim of this study was to show that the machine learning-based cancer cachexia classification model generalized well on the external validation cohort. METHODS This was a nationwide multicenter observational study conducted from October 2012 to April 2021 in China. Unsupervised consensus clustering analysis was applied based on demographic, anthropometric, nutritional, oncological, and quality-of-life data. Key characteristics of each cluster were identified using the standardized mean difference. We used logistic and Cox regression analysis to evaluate 1-, 3-, 5-y, and overall mortality. RESULTS A consensus clustering algorithm was performed for 4329 patients with cancer cachexia in the discovery cohort, and four clusters with distinct phenotypes were uncovered. From clusters 1 to 4, the clinical characteristics of patients showed a transition from almost unimpaired to mildly, moderately, and severely impaired. Consistently, an increase in mortality from clusters 1 to 4 was observed. The overall mortality rate was 32%, 40%, 54%, and 68%, and the median overall survival time was 21.9, 18, 16.7, and 13.6 mo for patients in clusters 1 to 4, respectively. Our machine learning-based model performed better in predicting mortality than the traditional model. External validation confirmed the above results. CONCLUSIONS Machine learning is valuable for phenotype classifications of patients with cancer cachexia. Detection of clinically distinct clusters among cachexic patients assists in scheduling personalized treatment strategies and in patient selection for clinical trials.
Collapse
Affiliation(s)
- Hao-Fan Wu
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang-Peng Yan
- Department of Automation, Tsinghua University, Beijing, China
| | - Qian Wu
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Yu
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hong-Xia Xu
- Department of Clinical Nutrition, Daping Hospital & Research Institute of Surgery, Third Military Medical University, Chongqing, China
| | - Chun-Hua Song
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Zeng-Qing Guo
- Department of Medical Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Wei Li
- Cancer Center of the First Hospital of Jilin University, Changchun, China
| | - Yan-Jun Xiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China; Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhe Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong, China
| | - Jie Luo
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Shu-Qun Cheng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Feng-Min Zhang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
5
|
McNearney TA, Digbeu BDE, Baillargeon JG, Ladnier D, Rahib L, Matrisian LM. Pre-Diagnosis Pain in Patients With Pancreatic Cancer Signals the Need for Aggressive Symptom Management. Oncologist 2023; 28:e1185-e1197. [PMID: 37285228 PMCID: PMC10712702 DOI: 10.1093/oncolo/oyad153] [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: 12/23/2022] [Accepted: 04/28/2023] [Indexed: 06/09/2023] Open
Abstract
OBJECTIVE This study assessed the impact of pancreatic cancer (PC) pain on associated symptoms, activities, and resource utilization from 2016 to 2020 in an online patient registry. PATIENTS AND METHODS Responses from PC patient volunteers (N = 1978) were analyzed from online surveys in a cross-sectional study. Comparisons were performed between PC patient groups reporting, (1) the presence vs. absence of pre-diagnosis PC pain, (2) high (4-8) vs. low (0-3) pain intensity scores on an 11-point numerical rating scale (NRS), and (3) year of PC diagnosis (2010-2020). Descriptive statistics and all bivariate analyses were performed using Chi-square or Fisher's Exact tests. RESULTS PC pain was the most frequently reported pre-diagnosis symptom (62%). Pre-diagnostic PC pain was reported more frequently by women, those with a younger age at diagnosis, and those with PC that spread to the liver and peritoneum. Those with pre-diagnostic PC pain vs. those without reported higher pain intensities (2.64 ± 2.54 vs.1.56 ± 2.01 NRS mean ± SD, respectively, P = .0039); increased frequencies of post-diagnosis symptoms of cramping after meals, feelings of indigestion, and weight loss (P = .02-.0001); and increased resource utilization in PC pain management: (ER visits N = 86 vs. N = 6, P = .018 and analgesic prescriptions, P < .03). The frequency of high pain intensity scores was not decreased over a recent 11-year span. CONCLUSIONS PC pain continues to be a prominent PC symptom. Patients reporting pre-diagnosis PC pain experience increased GI metastasis, symptoms burden, and are often undertreated. Its mitigation may require novel treatments, more resources dedicated to ongoing pain management and surveillance to improve outcomes.
Collapse
Affiliation(s)
- Terry A McNearney
- Scientific and Medical Affairs, Pancreatic Cancer Action Network (PanCAN), Manhattan Beach, CA, USA
| | | | | | - Dennis Ladnier
- Scientific and Medical Affairs, Pancreatic Cancer Action Network (PanCAN), Manhattan Beach, CA, USA
| | - Lola Rahib
- Scientific and Medical Affairs, Pancreatic Cancer Action Network (PanCAN), Manhattan Beach, CA, USA
| | - Lynn M Matrisian
- Scientific and Medical Affairs, Pancreatic Cancer Action Network (PanCAN), Manhattan Beach, CA, USA
| |
Collapse
|
6
|
Tao J, Zheng Y, Huang Q, Pu F, Shen Q, Hu Y. Patient-Reported Outcomes Measurement Information System in patients with gastrointestinal cancer: a scoping review. Support Care Cancer 2023; 31:567. [PMID: 37682320 DOI: 10.1007/s00520-023-08010-z] [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: 03/25/2023] [Accepted: 08/22/2023] [Indexed: 09/09/2023]
Abstract
PURPOSE The purpose of this study was to identify and evaluate the use of the Patient-Reported Outcomes Measurement Information System in patients with gastrointestinal cancer, as well as provide references for analyzing treatment outcomes, gauging prognostic risk, and assessing clinical symptoms and function in gastrointestinal cancer patients. METHODS PubMed, Web of Science, ProQuest, Embase, and CINAHL were systematically searched using keywords and controlled vocabulary through January 2023. RESULTS The scoping review collected 30 studies published from 2012 to 2022, including observational studies (n = 21), interventional studies (n = 4), and mixed studies (n = 5). Outcome indicators included mood, pain, fatigue, sleep, the ability to perform activities of daily living, social functioning, and other health problems. CONCLUSION Being a self-report questionnaire, the Patient-Reported Outcomes Measurement Information System offers a full assessment of patient's symptoms and quality of life, and accurately captures their actual thoughts, feelings, and experiences. Individuals with gastrointestinal cancer made frequent use of the Patient-Reported Outcomes Measurement Information System to provide an accurate measure of their health status.
Collapse
Affiliation(s)
- Jiaping Tao
- Zhejiang Chinese Medical University, Hangzhou, China
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yeping Zheng
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, China.
| | | | - Fengyan Pu
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qianqian Shen
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Yuanmin Hu
- The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
7
|
Aryal S, Bachman SL, Lyden K, Clay I. Measuring What Is Meaningful in Cancer Cachexia Clinical Trials: A Path Forward With Digital Measures of Real-World Physical Behavior. JCO Clin Cancer Inform 2023; 7:e2300055. [PMID: 37851933 PMCID: PMC10642875 DOI: 10.1200/cci.23.00055] [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: 04/03/2023] [Revised: 08/23/2023] [Accepted: 09/05/2023] [Indexed: 10/20/2023] Open
Abstract
PURPOSE The burden of cancer cachexia on patients' health-related quality of life, specifically their physical functioning, is well documented, but clinical trials thus far have failed to show meaningful improvement in physical functioning. The purpose of this review is to summarize existing methods of assessing physical function in cancer cachexia, outline a path forward for measuring what is meaningful to patients using digital measures derived from digital health technologies (DHTs), and discuss the current landscape of digital measures from the clinical and regulatory standpoint. DESIGN For this narrative review, peer-reviewed articles were searched on PubMed, clinical trials records were searched on clinicaltrials.gov, and records of digital measures submitted for regulatory qualification were searched on the US Food and Drug Administration's Drug Development Tool Qualification Program database. RESULTS There are gaps in assessing aspects of physical function that matter to patients. Existing assessment methods such as patient-reported outcomes and objective performance outcomes have limitations, including their episodic nature and burden to patients. DHTs such as wearable sensors can capture real-world physical behavior continuously, passively, and remotely, and may provide a more comprehensive picture of patients' everyday functioning. Recent regulatory submissions showcase potential clinical implementation of digital measures in various therapeutic areas. CONCLUSION Digital measures of real-world physical behavior present an opportunity to detect and demonstrate improvements in physical functioning in cancer cachexia, but evidence-based development is critical. For their use in clinical and regulatory decision making, studies demonstrating meaningfulness to patients as well as feasibility and validation are necessary.
Collapse
|
8
|
Cortez NE, Pathak S, Rodriguez Lanzi C, Hong BV, Crone R, Sule R, Wang F, Chen S, Gomes AV, Baar K, Mackenzie GG. A Ketogenic Diet in Combination with Gemcitabine Mitigates Pancreatic Cancer-Associated Cachexia in Male and Female KPC Mice. Int J Mol Sci 2023; 24:10753. [PMID: 37445930 PMCID: PMC10341838 DOI: 10.3390/ijms241310753] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/20/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Cancer-associated cachexia (CAC) is a critical contributor to pancreatic ductal adenocarcinoma (PDAC) mortality. Thus, there is an urgent need for new strategies to mitigate PDAC-associated cachexia; and the exploration of dietary interventions is a critical component. We previously observed that a ketogenic diet (KD) combined with gemcitabine enhances overall survival in the autochthonous LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx1-Cre (KPC) mouse model. In this study, we investigated the effect and cellular mechanisms of a KD in combination with gemcitabine on the maintenance of skeletal muscle mass in KPC mice. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD), a KD, a CD + gemcitabine (CG), or a KD + gemcitabine (KG) group. We observed that a KD or a KG-mitigated muscle strength declined over time and presented higher gastrocnemius weights compared CD-fed mice. Mechanistically, we observed sex-dependent effects of KG treatment, including the inhibition of autophagy, and increased phosphorylation levels of eIF2α in KG-treated KPC mice when compared to CG-treated mice. Our data suggest that a KG results in preservation of skeletal muscle mass. Additional research is warranted to explore whether this diet-treatment combination can be clinically effective in combating CAC in PDAC patients.
Collapse
Affiliation(s)
- Natalia E. Cortez
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Suraj Pathak
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Cecilia Rodriguez Lanzi
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Brian V. Hong
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
| | - Ryman Crone
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Rasheed Sule
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Fangyi Wang
- Department of Animal Science, University of California, One Shields Ave., Davis, CA 95616, USA;
| | - Shuai Chen
- Division of Biostatistics, Department of Public Health Sciences, University of California, One Shields Ave., Davis, CA 95616, USA;
- University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| | - Aldrin V. Gomes
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Keith Baar
- Department of Physiology and Membrane Biology, One Shields Ave., Davis, CA 95616, USA; (S.P.); (R.C.); (R.S.); (A.V.G.); (K.B.)
- Department of Neurobiology, Physiology and Behavior, University of California, One Shields Ave., Davis, CA 95616, USA
| | - Gerardo G. Mackenzie
- Department of Nutrition, University of California, One Shields Ave., Davis, CA 95616, USA; (N.E.C.); (C.R.L.); (B.V.H.)
- University of California Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA
| |
Collapse
|
9
|
Muranaka H, Hendifar A, Osipov A, Moshayedi N, Placencio-Hickok V, Tatonetti N, Stotland A, Parker S, Van Eyk J, Pandol SJ, Bhowmick NA, Gong J. Plasma Metabolomics Predicts Chemotherapy Response in Advanced Pancreatic Cancer. Cancers (Basel) 2023; 15:3020. [PMID: 37296982 PMCID: PMC10252041 DOI: 10.3390/cancers15113020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers. Developing biomarkers for chemotherapeutic response prediction is crucial for improving the dismal prognosis of advanced-PC patients (pts). To evaluate the potential of plasma metabolites as predictors of the response to chemotherapy for PC patients, we analyzed plasma metabolites using high-performance liquid chromatography-mass spectrometry from 31 cachectic, advanced-PC subjects enrolled into the PANCAX-1 (NCT02400398) prospective trial to receive a jejunal tube peptide-based diet for 12 weeks and who were planned for palliative chemotherapy. Overall, there were statistically significant differences in the levels of intermediates of multiple metabolic pathways in pts with a partial response (PR)/stable disease (SD) vs. progressive disease (PD) to chemotherapy. When stratified by the chemotherapy regimen, PD after 5-fluorouracil-based chemotherapy (e.g., FOLFIRINOX) was associated with decreased levels of amino acids (AAs). For gemcitabine-based chemotherapy (e.g., gemcitabine/nab-paclitaxel), PD was associated with increased levels of intermediates of glycolysis, the TCA cycle, nucleoside synthesis, and bile acid metabolism. These results demonstrate the feasibility of plasma metabolomics in a prospective cohort of advanced-PC patients for assessing the effect of enteral feeding as their primary source of nutrition. Metabolic signatures unique to FOLFIRINOX or gemcitabine/nab-paclitaxel may be predictive of a patient's response and warrant further study.
Collapse
Affiliation(s)
- Hayato Muranaka
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Andrew Hendifar
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Arsen Osipov
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Natalie Moshayedi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Veronica Placencio-Hickok
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nicholas Tatonetti
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Sarah Parker
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (A.S.); (S.P.); (J.V.E.)
| | - Stephen J. Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Research, VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Jun Gong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (H.M.); (A.H.); (A.O.); (N.M.); (V.P.-H.); (S.J.P.); (N.A.B.)
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
10
|
Ferrer M, Anthony TG, Ayres JS, Biffi G, Brown JC, Caan BJ, Cespedes Feliciano EM, Coll AP, Dunne RF, Goncalves MD, Grethlein J, Heymsfield SB, Hui S, Jamal-Hanjani M, Lam JM, Lewis DY, McCandlish D, Mustian KM, O'Rahilly S, Perrimon N, White EP, Janowitz T. Cachexia: A systemic consequence of progressive, unresolved disease. Cell 2023; 186:1824-1845. [PMID: 37116469 PMCID: PMC11059056 DOI: 10.1016/j.cell.2023.03.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 01/15/2023] [Accepted: 03/23/2023] [Indexed: 04/30/2023]
Abstract
Cachexia, a systemic wasting condition, is considered a late consequence of diseases, including cancer, organ failure, or infections, and contributes to significant morbidity and mortality. The induction process and mechanistic progression of cachexia are incompletely understood. Refocusing academic efforts away from advanced cachexia to the etiology of cachexia may enable discoveries of new therapeutic approaches. Here, we review drivers, mechanisms, organismal predispositions, evidence for multi-organ interaction, model systems, clinical research, trials, and care provision from early onset to late cachexia. Evidence is emerging that distinct inflammatory, metabolic, and neuro-modulatory drivers can initiate processes that ultimately converge on advanced cachexia.
Collapse
Affiliation(s)
- Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; MRC Cancer Unit, University of Cambridge, Hutchison Research Centre, Cambridge Biomedical Campus, Cambridge CB2 0XZ, UK
| | - Tracy G Anthony
- Department of Nutritional Sciences, Rutgers School of Environmental and Biological Sciences, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Janelle S Ayres
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Giulia Biffi
- University of Cambridge, Cancer Research UK Cambridge Institute, Li Ka Shing Centre, Cambridge CB2 0RE, UK
| | - Justin C Brown
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Bette J Caan
- Kaiser Permanente Northern California Division of Research, Oakland, CA 94612, USA
| | | | - Anthony P Coll
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Richard F Dunne
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Marcus D Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jonas Grethlein
- Ruprecht Karl University of Heidelberg, Heidelberg 69117, Germany
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Sheng Hui
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Mariam Jamal-Hanjani
- Department of Medical Oncology, University College London Hospitals, London WC1E 6DD, UK; Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - Jie Min Lam
- Cancer Research UK Lung Cancer Centre of Excellence and Cancer Metastasis Laboratory, University College London Cancer Institute, London WC1E 6DD, UK
| | - David Y Lewis
- The Beatson Institute for Cancer Research, Cancer Research UK, Glasgow G61 1BD, UK
| | - David McCandlish
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Karen M Mustian
- University of Rochester Medical Center, University of Rochester, Rochester, NY 14642, USA
| | - Stephen O'Rahilly
- Wellcome Trust-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eileen P White
- Rutgers Cancer Institute of New Jersey, Department of Molecular Biology and Biochemistry, Rutgers University, The State University of New Jersey, New Brunswick, NJ 08901, USA; Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Northwell Health Cancer Institute, Northwell Health, New Hyde Park, NY 11042, USA.
| |
Collapse
|
11
|
de Oliveira LC, Rosa KSDC, Gaspar T, Paiva BSR, Paiva CE, Peres WAF. Clinical usefulness of the Patient-Generated Subjective Global Assessment and modified Glasgow Prognostic Score in decision making concerning the indication of enteral nutritional therapy in patients with incurable cancer receiving palliative care. Nutrition 2023; 112:112057. [PMID: 37224572 DOI: 10.1016/j.nut.2023.112057] [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: 02/09/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/26/2023]
Abstract
OBJECTIVES This study aimed to identify the clinical usefulness of assessing nutritional status using validated tools for the indication of enteral nutrition for patients with incurable cancer in palliative care. METHODS In this prospective cohort study, patients were assessed for nutritional risk using the Patient-Generated Subjective Global Assessment and for cancer cachexia (CC) using the modified Glasgow Prognostic Score upon enrollment and after ∼30 d. The outcome was stable or improved Karnofsky Performance Status. Logistic regression models were used, providing the odds ratio (OR) and 95% confidence interval (CI). RESULTS A total of 180 patients participated. The only nutritional status parameter that was associated with function was CC. The less severe the CC, the more likely Karnofsky Performance Status was to remain stable or improve over 30 d (non-cachectic: OR = 1.95; 95% CI, 1.01-3.47; malnourished: OR = 1.06; 95% CI, 1.01-1.42). Furthermore, white skin color (OR = 1.79; 95% CI, 1.04-2.47), higher educational level (OR = 1.39; 95% CI, 1.13-2.78), and inadequate calorie intake (OR = 1.96; 95% CI, 1.02-2.81) were also associated with the outcome. CONCLUSIONS Using the modified Glasgow Prognostic Score to identify the existence and severity of CC, which is associated with function, has the potential to help clinical decision making concerning the indication of enteral nutrition in patients with incurable cancer receiving palliative care.
Collapse
Affiliation(s)
| | | | | | | | - Carlos Eduardo Paiva
- Research Group on Palliative Care and Quality of Life (GPQual), Barretos Cancer Hospital, Barretos, Brazil
| | | |
Collapse
|
12
|
Guo H, Zhang Y, Ma H, Gong P, Shi Y, Zhao W, Wang A, Liu M, Sun Z, Wang F, Wang Q, Ba X. T-stage-specific abdominal visceral fat, haematological nutrition indicators and inflammation as prognostic factors in patients with clear renal cell carcinoma. Adipocyte 2022; 11:133-142. [PMID: 35285399 PMCID: PMC8920171 DOI: 10.1080/21623945.2022.2048546] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Clear cell renal carcinoma (ccRCC) is the most common histological type of renal cancer and has the highest mortality. Several studies have been conducted on the relationship between adipose tissue and ccRCC prognosis, however, the results have been inconsistent to date. The current study aimed at establishing a link between abdominal fat composition and short-term prognosis in patients with ccRCC after T-stage stratification. We retrospectively analysed 250 patients with pathologically confirmed ccRCC (173 low T-stage and 77 high T-stage) in our hospital. The computed tomography (CT) images were evaluated using ImageJ. Then, subcutaneous and visceral fat areas (SFA and VFA), total fat areas (TFA) and the relative VFA (rVFA) were measured and computed. Meanwhile, biochemical indices of blood serum were analysed. The results showed that rVFA in low T-stage cohort who had a history of short-term postoperative complications were significantly lower than those who did not. No such association was observed in the high T-stage cohort. Further investigation revealed that the correlations between biochemical indexes and fat area-related variables varied across T-stage groups. As a result, rVFA is a reliable independent predictor of short-term prognosis in patients with low T-stage ccRCC but not in patients with high T-stage ccRCC.
Collapse
Affiliation(s)
- Hao Guo
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Yumei Zhang
- Department of Radiology, Lanshan Branch of Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Heng Ma
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Peiyou Gong
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Yinghong Shi
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Wenlei Zhao
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Aijie Wang
- Department of Radiology, Yaitai Shan Hospital, Shandong province, Yantai, China
| | - Ming Liu
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Zehua Sun
- Department of Radiology, Yantai Yuhuangding Hospital, Shandong province, Yantai, China
| | - Fang Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
| | - Qing Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong province, Jinan, China
| | - Xinru Ba
- Department of Radiology, Yaitai Shan Hospital, Shandong province, Yantai, China
| |
Collapse
|
13
|
Wang Y, An Z, Lin D, Jin W. Targeting cancer cachexia: Molecular mechanisms and clinical study. MedComm (Beijing) 2022; 3:e164. [PMID: 36105371 PMCID: PMC9464063 DOI: 10.1002/mco2.164] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/01/2022] [Accepted: 07/07/2022] [Indexed: 11/12/2022] Open
Abstract
Cancer cachexia is a complex systemic catabolism syndrome characterized by muscle wasting. It affects multiple distant organs and their crosstalk with cancer constitute cancer cachexia environment. During the occurrence and progression of cancer cachexia, interactions of aberrant organs with cancer cells or other organs in a cancer cachexia environment initiate a cascade of stress reactions and destroy multiple organs including the liver, heart, pancreas, intestine, brain, bone, and spleen in metabolism, neural, and immune homeostasis. The role of involved organs turned from inhibiting tumor growth into promoting cancer cachexia in cancer progression. In this review, we depicted the complicated relationship of cancer cachexia with the metabolism, neural, and immune homeostasis imbalance in multiple organs in a cancer cachexia environment and summarized the treatment progress in recent years. And we discussed the molecular mechanism and clinical study of cancer cachexia from the perspective of multiple organs metabolic, neurological, and immunological abnormalities. Updated understanding of cancer cachexia might facilitate the exploration of biomarkers and novel therapeutic targets of cancer cachexia.
Collapse
Affiliation(s)
- Yong‐Fei Wang
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Zi‐Yi An
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| | - Dong‐Hai Lin
- Key Laboratory for Chemical Biology of Fujian ProvinceMOE Key Laboratory of Spectrochemical Analysis and InstrumentationCollege of Chemistry and Chemical EngineeringXiamen UniversityXiamenChina
| | - Wei‐Lin Jin
- The First Clinical Medical College of Lanzhou UniversityLanzhouChina
- Institute of Cancer NeuroscienceMedical Frontier Innovation Research CenterThe First Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
14
|
Adipose Tissue Wasting as a Determinant of Pancreatic Cancer-Related Cachexia. Cancers (Basel) 2022; 14:cancers14194754. [PMID: 36230682 PMCID: PMC9563866 DOI: 10.3390/cancers14194754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 12/03/2022] Open
Abstract
Simple Summary Pancreatic cancer (PC) is one of the deadliest cancers in the US. The poor prognosis of PC is related to diagnostic delay and the presence of unintended weight loss (cachexia) that commonly presents in PC patients even before diagnosis. However, the current understanding of how PC mediates cachexia is limited, and there are few treatments clinically available for cachexia. Based on the current literature, we demonstrate that PC-related cachexia primarily results from the wasting of adipose tissue, once thought to be merely a storage depot but now appreciated as an instrumental metabolic organ in the body. In addition, poor survival in PC patients was found to be associated with adipose tissue loss at diagnosis and during treatment. Therefore, identifying potential mediators and molecular mechanisms underlying adipose tissue loss would promise to pave the way for the development of effective interventions for PC-related cachexia Abstract Pancreatic cancer (PC) is the third leading cause of cancer-related death in the US, and its 5-year survival rate is approximately 10%. The low survival rates largely stem from diagnostic delay and the presence of significant adipose tissue and muscle wasting, commonly referred to as cachexia. Cachexia is present in nearly 80% of PC patients and is a key cause of poor response to treatment and about 20% of death in PC patients. However, there are few clinical interventions proven to be effective against PC-related cachexia. Different cancer types feature distinct secretome profiles and functional characteristics which would lead to cachexia development differently. Therefore, here we discuss affected tissues and potential mechanisms leading to cachexia in PC. We postulate that the most affected tissue during the development of PC-related cachexia is adipose tissue, historically and still thought to be just an inert repository for excess energy in relation to cancer-related cachexia. Adipose tissue loss is considerably greater than muscle loss in quantity and shows a correlation with poor survival in PC patients. Moreover, we suggest that PC mediates adipose atrophy by accelerating adipocyte lipid turnover and fibroblast infiltration.
Collapse
|
15
|
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
|
16
|
Gong J, Thomassian S, Kim S, Gresham G, Moshayedi N, Ye JY, Yang JC, Jacobs JP, Lo S, Nissen N, Gaddam S, Tighiouart M, Osipov A, Hendifar A. Phase I trial of Bermekimab with nanoliposomal irinotecan and 5-fluorouracil/folinic acid in advanced pancreatic ductal adenocarcinoma. Sci Rep 2022; 12:15013. [PMID: 36056179 PMCID: PMC9440135 DOI: 10.1038/s41598-022-19401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/29/2022] [Indexed: 11/09/2022] Open
Abstract
In this phase I dose-escalation trial, we assess the maximum tolerated dose (MTD) of Bermekimab in combination with Nanoliposomal Irinotecan (Nal-Iri) and 5-Fluorouracil/Folinic Acid (5-FU/FA). Secondarily, we investigate effects on weight, lean body mass, quality-of-life, the gut microbiome composition, inflammatory biomarkers, progression-free survival, and overall survival. This was a single-arm, open-label adaptive Bayesian dose-escalation study of Bermekimab combined with Nal-Iri and 5FU/FA in patients with advanced or locally advanced PDAC who failed gemcitabine-based chemotherapy. 22 patients enrolled between 2017 and 2019. 3 of 21 patients experienced dose-limiting toxicities attributable to the chemotherapy backbone. 58% (10/17) of patients exhibited weight stability. Physical performance status was preserved among all subjects. Patients reported improvements in quality-of-life metrics via QLQ-PAN26 questioner (-3.6, p = 0.18) and functional well-being (1.78, p = 0.02). Subjects exhibited a decrease in inflammatory cytokines, notably, vascular endothelial growth factor (-0.86, p = 0.017) with Bermekimab. Bermekimab treatment was associated with an increased abundance of gut health-promoting bacterial genera Akkermansia, with 3.82 Log2-fold change from baseline. In sum, Bermekimab is safe to be used in conjunction with Nal-Iri and 5-FU/FA chemotherapy. This benign toxicological profile warrants further Phase I/II investigation of Bermekimab in combinatorial strategies, and the impact of anti-IL-1α antibodies on the gut microbiome.Clinical trials registration: NCT03207724 05/07/2017.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Shant Thomassian
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Sungjin Kim
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Gillian Gresham
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Natalie Moshayedi
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Jason Y Ye
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Julianne C Yang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA
| | - Simon Lo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Nick Nissen
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Srinivas Gaddam
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Mourad Tighiouart
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Arsen Osipov
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Andrew Hendifar
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA.
| |
Collapse
|
17
|
Law ML. Cancer cachexia: Pathophysiology and association with cancer-related pain. FRONTIERS IN PAIN RESEARCH 2022; 3:971295. [PMID: 36072367 PMCID: PMC9441771 DOI: 10.3389/fpain.2022.971295] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Cachexia is a syndrome of unintentional body weight loss and muscle wasting occurring in 30% of all cancer patients. Patients with cancers most commonly leading to brain metastases have a risk for cachexia development between 20 and 80%. Cachexia causes severe weakness and fatigue and negatively impacts quality and length of life. The negative energy balance in cachectic patients is most often caused by a combination of increased energy expenditure and decreased energy intake. Basal metabolic rate may be elevated due to tumor secreted factors and a systemic inflammatory response leading to inefficiency in energy production pathways and increased energy demand by the tumor and host tissues. A growing body of research explores physiological and molecular mechanisms of metabolic dysregulation in cachexia. However, decreased energy intake and physical functioning also remain important contributors to cachexia pathogenesis. Pain associated with metastatic malignancy is significantly associated with inflammation, thus making inflammation a common link between cancer pain and cachexia. Pain may also influence appetite and food intake and exacerbate fatigue and functional decline, potentially contributing to cachexia severity. Cancer pain and cachexia often occur simultaneously; however, causal relationships remain to be established. Appropriate assessment and treatment of pain in advanced cancer patients may positively impact nutrition status and physical functioning, slowing the progression of cachexia and improving quality and length of life for patients.
Collapse
|
18
|
Hendifar A, Akinsola R, Muranaka H, Osipov A, Thomassian S, Moshayedi N, Yang J, Jacobs J, Devkota S, Bhowmick N, Gong J. Gut microbiome and pancreatic cancer cachexia: An evolving relationship. World J Gastrointest Oncol 2022; 14:1218-1226. [PMID: 36051103 PMCID: PMC9305570 DOI: 10.4251/wjgo.v14.i7.1218] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/10/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
Nearly 80% of patients with pancreatic ductal adenocarcinoma (PDAC) develop cachexia along their disease course. Cachexia is characterized by progressive weight loss, muscle wasting, and systemic inflammation and has been linked to poorer outcomes and impairments in quality of life. Management of PDAC cachexia has historically involved a multidisciplinary effort comprised of nutritional support, pancreatic enzyme replacement therapy, and/or pharmacologic interventions. Despite current interventions to mitigate PDAC cachexia, a significant proportion of patients continue to die from complications associated with cachexia underscoring the need for novel insights and treatments for this syndrome. We highlight the feasibility and effectiveness of a recent enteral feeding prospective trial at our institution to improve cachexia outcomes in patients with advanced PDAC. Additionally, we were among the first to characterize the stool microbiome composition in patients with advanced PDAC receiving enteral feeding for the treatment of cachexia. Novel insights into the relationship between enteral nutritional support, cachexia, and the gut microbiome are presented. These promising results are discussed in the context of a potential ability to modulate the stool microbiome as a new interventional strategy to mitigate PDAC cachexia.
Collapse
Affiliation(s)
- Andrew Hendifar
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Rasaq Akinsola
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Hayato Muranaka
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Arsen Osipov
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Shant Thomassian
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Natalie Moshayedi
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Julianne Yang
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, University of California, Los Angeles, CA 90095, United States
| | - Jonathan Jacobs
- Department of Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, United States
| | - Suzanne Devkota
- Cedars-Sinai Medical Center, Department of Medicine, Inflammatory Bowel and Immunobiology Research Institute, University of California, Los Angeles, CA 90048, United States
| | - Neil Bhowmick
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Jun Gong
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, United States
| |
Collapse
|