1
|
Chen LN, Ma X, Herzberg B, Henick BS, Biswas AK, Acharyya S, Shu CA. Weight loss in patients on osimertinib for metastatic EGFR-mutant non-small cell lung cancer. Oncologist 2024:oyae315. [PMID: 39703162 DOI: 10.1093/oncolo/oyae315] [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: 02/12/2024] [Accepted: 09/21/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Cachexia is characterized by weight loss and decline in muscle mass and function and is a poor prognostic factor among patients with cancer. Patients with metastatic EGFR-mutant non-small cell lung cancer (NSCLC) derive remarkable survival benefits with osimertinib, a third-generation EGFR tyrosine kinase inhibitor. It is not known whether patients treated with osimertinib experience any weight loss or whether weight loss impacts patient outcomes. Therefore, we sought to describe the frequency and consequences of weight loss in this patient population. MATERIALS AND METHODS We conducted a single-center retrospective pilot study of 56 patients treated with first-line osimertinib for metastatic EGFR-mutant NSCLC. We defined on-treatment weight loss as a loss of ≥5% body weight at 6 or 12 months of treatment. We described the characteristics of patients with and without on-treatment weight loss and differences in progression-free survival (PFS), time on treatment with osimertinib, and overall survival (OS). RESULTS Forty-six percent (n = 26) of patients met the criteria for on-treatment weight loss. There were no significant differences in patient or disease characteristics between patients with and without weight loss. Compared to patients without weight loss, patients with weight loss had similar PFS and time on treatment with osimertinib. Yet, patients with weight loss had significantly worse overall survival (HR 4.91, 95% CI, 1.56-15.5, P = .007). CONCLUSION Weight loss was observed in nearly half of patients with metastatic EGFR-mutant NSCLC treated with osimertinib, and patients with weight loss had significantly worse overall survival.
Collapse
Affiliation(s)
- Lanyi Nora Chen
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
| | - Xin Ma
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
- Department of Statistics, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Benjamin Herzberg
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
| | - Brian S Henick
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
| | - Anup K Biswas
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
- Institute for Cancer Genetics and Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Swarnali Acharyya
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
- Institute for Cancer Genetics and Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY 10032, United States
| | - Catherine A Shu
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center New York, NY 10032, United States
| |
Collapse
|
2
|
Park MA, Whelan CJ, Ahmed S, Boeringer T, Brown J, Crowder SL, Gage K, Gregg C, Jeong DK, Jim HSL, Judge AR, Mason TM, Parker N, Pillai S, Qayyum A, Rajasekhara S, Rasool G, Tinsley SM, Schabath MB, Stewart P, West J, McDonald P, Permuth JB. Defining and Addressing Research Priorities in Cancer Cachexia through Transdisciplinary Collaboration. Cancers (Basel) 2024; 16:2364. [PMID: 39001427 PMCID: PMC11240731 DOI: 10.3390/cancers16132364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
For many patients, the cancer continuum includes a syndrome known as cancer-associated cachexia (CAC), which encompasses the unintended loss of body weight and muscle mass, and is often associated with fat loss, decreased appetite, lower tolerance and poorer response to treatment, poor quality of life, and reduced survival. Unfortunately, there are no effective therapeutic interventions to completely reverse cancer cachexia and no FDA-approved pharmacologic agents; hence, new approaches are urgently needed. In May of 2022, researchers and clinicians from Moffitt Cancer Center held an inaugural retreat on CAC that aimed to review the state of the science, identify knowledge gaps and research priorities, and foster transdisciplinary collaborative research projects. This review summarizes research priorities that emerged from the retreat, examples of ongoing collaborations, and opportunities to move science forward. The highest priorities identified include the need to (1) evaluate patient-reported outcome (PRO) measures obtained in clinical practice and assess their use in improving CAC-related outcomes; (2) identify biomarkers (imaging, molecular, and/or behavioral) and novel analytic approaches to accurately predict the early onset of CAC and its progression; and (3) develop and test interventions (pharmacologic, nutritional, exercise-based, and through mathematical modeling) to prevent CAC progression and improve associated symptoms and outcomes.
Collapse
Affiliation(s)
- Margaret A. Park
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Christopher J. Whelan
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Sabeen Ahmed
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Tabitha Boeringer
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Joel Brown
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Sylvia L. Crowder
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Kenneth Gage
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Christopher Gregg
- School of Medicine, University of Utah, Salt Lake City, UT 84113, USA;
| | - Daniel K. Jeong
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Heather S. L. Jim
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Andrew R. Judge
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA;
| | - Tina M. Mason
- Department of Nursing Research, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Nathan Parker
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
| | - Smitha Pillai
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (T.B.); (S.P.)
| | - Aliya Qayyum
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (K.G.); (D.K.J.); (A.Q.)
| | - Sahana Rajasekhara
- Department of Supportive Care Medicine, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Ghulam Rasool
- Department of Machine Learning, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.A.); (G.R.)
| | - Sara M. Tinsley
- Department of Health Outcomes and Behavior, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (S.L.C.); (H.S.L.J.); (N.P.); (S.M.T.)
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Matthew B. Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Paul Stewart
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| | - Jeffrey West
- Department of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; (J.B.); (J.W.)
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Patricia McDonald
- Department of Metabolism and Cancer Physiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Lexicon Pharmaceuticals, Inc., Woodlands, TX 77381, USA
| | - Jennifer B. Permuth
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA;
| |
Collapse
|
3
|
Hasuo H, Mori K, Matsuoka H, Sakuma H, Ishikawa H. An Estimation Formula for Resonance Frequency Using Sex and Height for Healthy Individuals and Patients with Incurable Cancers. Appl Psychophysiol Biofeedback 2024; 49:125-132. [PMID: 37702816 PMCID: PMC10869367 DOI: 10.1007/s10484-023-09602-5] [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] [Accepted: 07/25/2023] [Indexed: 09/14/2023]
Abstract
Resonance frequency breathing is a technique that involves breathing that maximizes heart rate variability. It is specific to individuals and is determined through a procedure taking approximately 30 min, using a procedure that is often best carried out at specialized medical institutions. This is a physical and time-consuming burden because of hospital visits and measurements, particularly for patients with cancer. Therefore it would be beneficial if a procedure can be found to determine resonance frequency from the patient's physical characteristics, without the need for special assessment procedures. This exploratory cross-sectional study examined the correlation between individual characteristics and resonance frequency in healthy volunteers. Multiple regression analysis was performed with the measured resonance frequency as the target variable and individual characteristic parameters as explanatory variables. The study aims to build an estimation formula for resonance frequency with some of these parameters and assess its validity. In addition, the validity of the formula's applicability to patients with incurable cancers is assessed. A total of 122 healthy volunteers and 32 patients with incurable cancers were recruited as participants. The median resonance frequency of 154 participants was six breaths per min. Sex and height were selected as explanatory variables associated with the measured resonance frequency in the volunteers. The estimation formula for resonance frequency using individual characteristics was 17.90-0.07 × height for men and 15.88-0.06 × height for women. Adjusted R-squared values were 0.55 for men and 0.47 for women. When the measured resonance frequency in patients with incurable cancers was six breaths per minute or less, the resonance frequency estimated by this formula was slightly larger than the measured ones. Information on individual characteristics, such as sex and height, which can be easily obtained, was useful to construct an estimation formula for resonance frequency.
Collapse
Affiliation(s)
- Hideaki Hasuo
- Department of Psychosomatic Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, 573-1090, Japan.
| | - Keita Mori
- Clinical Research Support Center, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiromichi Matsuoka
- Department of Psycho-Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroko Sakuma
- Department of Psychosomatic Medicine, Kansai Medical University, Shinmachi 2-5-1, Hirakata, Osaka, 573-1090, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
4
|
Ispoglou T, McCullough D, Windle A, Nair S, Cox N, White H, Burke D, Kanatas A, Prokopidis K. Addressing cancer anorexia-cachexia in older patients: Potential therapeutic strategies and molecular pathways. Clin Nutr 2024; 43:552-566. [PMID: 38237369 DOI: 10.1016/j.clnu.2024.01.009] [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: 11/03/2022] [Revised: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
Cancer cachexia (CC) syndrome, a feature of cancer-associated muscle wasting, is particularly pronounced in older patients, and is characterised by decreased energy intake and upregulated skeletal muscle catabolic pathways. To address CC, appetite stimulants, anabolic drugs, cytokine mediators, essential amino acid supplementation, nutritional counselling, cognitive behavioural therapy, and enteral nutrition have been utilised. However, pharmacological treatments that have also shown promising results, such as megestrol acetate, anamorelin, thalidomide, and delta-9-tetrahydrocannabinol, have been associated with gastrointestinal and cardiovascular complications. Emerging evidence on the efficacy of probiotics in modulating gut microbiota also presents a promising adjunct to traditional therapies, potentially enhancing nutritional absorption and systemic inflammation control. Additionally, low-dose olanzapine has demonstrated improved appetite and weight management in older patients undergoing chemotherapy, offering a potential refinement to current therapeutic approaches. This review aims to elucidate the molecular mechanisms underpinning CC, with a particular focus on the role of anorexia in exacerbating muscle wasting, and to propose pharmacological and non-pharmacological strategies to mitigate this syndrome, particularly emphasising the needs of an older demographic. Future research targeting CC should focus on refining appetite-stimulating drugs with fewer side-effects, specifically catering to the needs of older patients, and investigating nutritional factors that can either enhance appetite or minimise suppression of appetite in individuals with CC, especially within this vulnerable group.
Collapse
Affiliation(s)
| | | | - Angela Windle
- Department of Nursing and Midwifery, School of Human and Health Sciences, University of Huddersfield, Huddersfield, UK; School of Medicine, University of Leeds, Leeds, UK
| | | | - Natalie Cox
- Academic Geriatric Medicine, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Helen White
- School of Health, Leeds Beckett University, Leeds, UK
| | - Dermot Burke
- School of Medicine, University of Leeds, Leeds, UK
| | | | - Konstantinos Prokopidis
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK; Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool, UK
| |
Collapse
|
5
|
Souza-Silva RD, Calixto-Lima L, Varea Maria Wiegert E, de Oliveira LC. Decision tree algorithm to predict mortality in incurable cancer: a new prognostic model. BMJ Support Palliat Care 2024:spcare-2023-004581. [PMID: 38242639 DOI: 10.1136/spcare-2023-004581] [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: 08/30/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
OBJECTIVES To develop and validate a new prognostic model to predict 90-day mortality in patients with incurable cancer. METHODS In this prospective cohort study, patients with incurable cancer receiving palliative care (n = 1322) were randomly divided into two groups: development (n = 926, 70%) and validation (n = 396, 30%). A decision tree algorithm was used to develop a prognostic model with clinical variables. The accuracy and applicability of the proposed model were assessed by the C-statistic, calibration and receiver operating characteristic (ROC) curve. RESULTS Albumin (75.2%), C reactive protein (CRP) (47.7%) and Karnofsky Performance Status (KPS) ≥50% (26.5%) were the variables that most contributed to the classification power of the prognostic model, named Simple decision Tree algorithm for predicting mortality in patients with Incurable Cancer (acromion STIC). This was used to identify three groups of increasing risk of 90-day mortality: STIC-1 - low risk (probability of death: 0.30): albumin ≥3.6 g/dL, CRP <7.8 mg/dL and KPS ≥50%; STIC-2 - medium risk (probability of death: 0.66 to 0.69): albumin ≥3.6 g/dL, CRP <7.8 mg/dL and KPS <50%, or albumin ≥3.6 g/dL and CRP ≥7.8 mg/dL; STIC-3 - high risk (probability of death: 0.79): albumin <3.6 g/dL. In the validation dataset, good accuracy (C-statistic ≥0.71), Hosmer-Lemeshow p=0.12 and area under the ROC curve=0.707 were found. CONCLUSIONS STIC is a valid, practical tool for stratifying patients with incurable cancer into three risk groups for 90-day mortality.
Collapse
|
6
|
Malta FAPS, Gonçalves DC. A triple-masked, two-center, randomized parallel clinical trial to assess the superiority of eight weeks of grape seed flour supplementation against placebo for weight loss attenuation during perioperative period in patients with cachexia associated with colorectal cancer: a study protocol. Front Endocrinol (Lausanne) 2024; 14:1146479. [PMID: 38313843 PMCID: PMC10834683 DOI: 10.3389/fendo.2023.1146479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 12/07/2023] [Indexed: 02/06/2024] Open
Abstract
Background Progressive, involuntary weight and lean mass loss in cancer are linked to cachexia, a prevalent syndrome in gastrointestinal malignancies that impacts quality of life, survival and postoperative complications. Its pathophysiology is complex and believed to involve proinflammatory cytokine-mediated systemic inflammation resulting from tumor-host interaction, oxidative stress, abnormal metabolism and neuroendocrine changes. Therapeutic options for cachexia remain extremely limited, highlighting the need for clinical research targeting new interventions. Thus, this study primarily assesses the effects of grape-seed flour (GSF), rich in polyphenols and fibers, for attenuating perioperative weight loss in colorectal cancer. Methods This is a dual-center, triple-masked, placebo-controlled, parallel-group, phase II, randomized clinical trial designed to investigate GSF supplementation in subjects with pre- or cachexia associated with colorectal cancer during the perioperative period. Eighty-two participants will receive 8g of GSF or cornstarch (control) for 8 weeks. Assessments are scheduled around surgery: pre-intervention (4 weeks prior), day before, first week after, and post-intervention (4 weeks later). The primary endpoint is the difference in body weight mean change from baseline to week 8. The secondary endpoints describe the harms from 8-week supplementation and assess its superiority to improve body composition, post-surgical complications, quality of life, anorexia, fatigue, gastrointestinal symptoms, and handgrip strength. The study will also explore its effects on gut bacteria activity and composition, systemic inflammation, and muscle metabolism. Discussion The current trial addresses a gap within the field of cancer cachexia, specifically focusing on the potential role of a nutritional intervention during the acute treatment phase. GSF is expected to modulate inflammation and oxidative stress, both involved in muscle and intestinal dysfunction. The research findings hold substantial implications for enhancing the understanding about cachexia pathophysiology and may offer a new clinical approach to managing cachexia at a critical point in treatment, directly impacting clinical outcomes. Trial registration The Brazilian Registry of Clinical Trials (ReBEC), RBR-5p6nv8b; UTN: U1111-1285-9594. Prospectively registered on February 07, 2023.
Collapse
|
7
|
Blum D, Vagnildhaug OM, Stene GB, Maddocks M, Sørensen J, Laird BJA, Prado CM, Skeidsvoll Solheim T, Arends J, Hopkinson J, Jones CA, Schlögl M. Top Ten Tips Palliative Care Clinicians Should Know About Cachexia. J Palliat Med 2023; 26:1133-1138. [PMID: 36723498 DOI: 10.1089/jpm.2022.0598] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Cachexia is a multifactorial syndrome that is common in cancer and chronic disease. It is often underdiagnosed and therefore goes untreated or undertreated. Cachexia causes suffering across biopsychosocial domains and affects patients and their loved ones. In this article, a group of clinicians and researchers across cancer care, nutrition, and exercise offers tips about assessment, classification, and management of cachexia, with attention to its stage. The required multimodal management of cachexia mirrors well the interprofessional collaboration that is the mainstay of interdisciplinary palliative care and attention to screening, diagnosis, and management of cachexia is critical to maximize patients' quality of life.
Collapse
Affiliation(s)
- David Blum
- Competence Center for Palliative Care, Department of Radiation Oncology, University Hospital Zurich, and University of Zurich UZH, Zurich, Switzerland
| | - Ola Magne Vagnildhaug
- European Palliative Care Research Centre (PRC), Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology and Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Guro Birgitte Stene
- Faculty of Medicine and Health Science, Department of Neuromedicine and Movement Science, The Norwegian University of Science and Technology, Trondheim, Norway
| | - Matthew Maddocks
- Cicely Saunders Institute of Palliative Care, Policy and Rehabilitation, King's College London, London, United Kingdom
| | - Jonas Sørensen
- Department of Biomedical Sciences, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Centre for Cancer and Organ Diseases, Rigshospitalet, Denmark
| | - Barry J A Laird
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Carla M Prado
- Division of Human Nutrition, Department of Agricultural, Food and Nutritional Science, University of Alberta, Alberta, Canada
| | - Tora Skeidsvoll Solheim
- Cancer Clinic, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
- Faculty of Medicine and Health Sciences, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jann Arends
- Department of Medicine I, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jane Hopkinson
- School of Healthcare Sciences, Cardiff University, Cardiff, United Kingdom
| | - Christopher A Jones
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mathias Schlögl
- Division of Geriatric Medicine, Clinic Barmelweid, Barmelweid, Switzerland
| |
Collapse
|
8
|
Dhiman P, Ma J, Andaur Navarro CL, Speich B, Bullock G, Damen JAA, Hooft L, Kirtley S, Riley RD, Van Calster B, Moons KGM, Collins GS. Overinterpretation of findings in machine learning prediction model studies in oncology: a systematic review. J Clin Epidemiol 2023; 157:120-133. [PMID: 36935090 DOI: 10.1016/j.jclinepi.2023.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVES In biomedical research, spin is the overinterpretation of findings, and it is a growing concern. To date, the presence of spin has not been evaluated in prognostic model research in oncology, including studies developing and validating models for individualized risk prediction. STUDY DESIGN AND SETTING We conducted a systematic review, searching MEDLINE and EMBASE for oncology-related studies that developed and validated a prognostic model using machine learning published between 1st January, 2019, and 5th September, 2019. We used existing spin frameworks and described areas of highly suggestive spin practices. RESULTS We included 62 publications (including 152 developed models; 37 validated models). Reporting was inconsistent between methods and the results in 27% of studies due to additional analysis and selective reporting. Thirty-two studies (out of 36 applicable studies) reported comparisons between developed models in their discussion and predominantly used discrimination measures to support their claims (78%). Thirty-five studies (56%) used an overly strong or leading word in their title, abstract, results, discussion, or conclusion. CONCLUSION The potential for spin needs to be considered when reading, interpreting, and using studies that developed and validated prognostic models in oncology. Researchers should carefully report their prognostic model research using words that reflect their actual results and strength of evidence.
Collapse
Affiliation(s)
- Paula Dhiman
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Jie Ma
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Constanza L Andaur Navarro
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Benjamin Speich
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK; Meta-Research Centre, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Garrett Bullock
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Johanna A A Damen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lotty Hooft
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Shona Kirtley
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Richard D Riley
- Centre for Prognosis Research, School of Medicine, Keele University, Staffordshire, UK, ST5 5BG
| | - Ben Van Calster
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium; Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands; EPI-centre, KU Leuven, Leuven, Belgium
| | - Karel G M Moons
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gary S Collins
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
9
|
Tan S, Xu J, Wang J, Zhang Z, Li S, Yan M, Tang M, Liu H, Zhuang Q, Xi Q, Meng Q, Jiang Y, Wu G. Development and validation of a cancer cachexia risk score for digestive tract cancer patients before abdominal surgery. J Cachexia Sarcopenia Muscle 2023; 14:891-902. [PMID: 36880286 PMCID: PMC10067494 DOI: 10.1002/jcsm.13207] [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: 07/23/2022] [Revised: 10/26/2022] [Accepted: 02/02/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Cancer cachexia is prevalent in digestive tract cancer patients and has significant impacts on prognosis; it is vital to identify individuals who are at risk of cancer cachexia to allow for appropriate evaluation and treatment. This study evaluated whether digestive tract cancer patients with a risk of cancer cachexia and who had a risk of adverse survival could be identified before abdominal surgery. METHODS This large-scale cohort study involved patients who underwent abdominal surgery between January 2015 and December 2020 to treat digestive tract cancer. Participants were allocated to the development cohort, the validation cohort, or the application cohort. Univariate and multivariate analyses of the development cohort were performed to detect distinct risk variables for cancer cachexia to create a cancer cachexia risk score. The performance of the risk score across all the three cohorts was assessed through calculating the area under the receiver operating characteristic curve (AUC), as well as calibration and decision curves. We tested how well the score predicted survival outcomes in the application cohort. RESULTS A total of 16 264 patients (median 64 years of age; 65.9% male) were included, with 8743 in the development cohort, 5828 in the validation cohort, and 1693 in the application cohort. Seven variables were identified as independent predictive factors and were included in the cancer cachexia risk score: cancer site, cancer stage, time from symptom onset to hospitalization, appetite loss, body mass index, skeletal muscle index, and neutrophil-lymphocyte ratio. The risk score predicting cancer cachexia owns a good discrimination, with the mean AUC of 0.760 (P < 0.001) in the development cohort, 0.743 (P < 0.001) in the validation cohort, and 0.751 (P < 0.001) in the application cohort, respectively, and had an excellent calibration (all P > 0.05). The decision curve analysis revealed net benefits of the risk score across a range of risk thresholds in the three cohorts. In the application cohort, compared with the high-risk group, the low-risk group experienced significantly longer overall survival [hazard ratio (HR) 2.887, P < 0.001] as well as relapse-free survival (HR 1.482, P = 0.01). CONCLUSIONS The cancer cachexia risk score constructed and validated demonstrated good performance in identifying those digestive tract cancer patients before abdominal surgery at a higher risk of cancer cachexia and unfavourable survival. This risk score can help clinicians to enhance their capabilities to screen for cancer cachexia, assess patient prognosis, and strengthen early decision-making on targeted approaches to attune cancer cachexia for digestive tract cancer patients before abdominal surgery.
Collapse
Affiliation(s)
- Shanjun Tan
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiahao Xu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junjie Wang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhige Zhang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuhao Li
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mingyue Yan
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min Tang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Liu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiulin Zhuang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiulei Xi
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinyang Meng
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Jiang
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guohao Wu
- Department of General Surgery/Shanghai Clinical Nutrition Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Cury SS, de Moraes D, Oliveira JS, Freire PP, dos Reis PP, Batista ML, Hasimoto ÉN, Carvalho RF. Low muscle mass in lung cancer is associated with an inflammatory and immunosuppressive tumor microenvironment. J Transl Med 2023; 21:116. [PMID: 36774484 PMCID: PMC9921698 DOI: 10.1186/s12967-023-03901-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/17/2023] [Indexed: 02/13/2023] Open
Abstract
BACKGROUND Computed tomographies (CT) are useful for identifying muscle loss in non-small lung cancer (NSCLC) cachectic patients. However, we lack consensus on the best cutoff point for pectoralis muscle loss. We aimed to characterize NSCLC patients based on muscularity, clinical data, and the transcriptional profile from the tumor microenvironment to build a cachexia classification model. METHODS We used machine learning to generate a muscle loss prediction model, and the tumor's cellular and transcriptional profile was characterized in patients with low muscularity. First, we measured the pectoralis muscle area (PMA) of 211 treatment-naive NSCLC patients using CT available in The Cancer Imaging Archive. The cutoffs were established using machine learning algorithms (CART and Cutoff Finder) on PMA, clinical, and survival data. We evaluated the prediction model in a validation set (36 NSCLC). Tumor RNA-Seq (GSE103584) was used to profile the transcriptome and cellular composition based on digital cytometry. RESULTS CART demonstrated that a lower PMA was associated with a high risk of death (HR = 1.99). Cutoff Finder selected PMA cutoffs separating low-muscularity (LM) patients based on the risk of death (P-value = 0.003; discovery set). The cutoff presented 84% of success in classifying low muscle mass. The high risk of LM patients was also found in the validation set. Tumor RNA-Seq revealed 90 upregulated secretory genes in LM that potentially interact with muscle cell receptors. The LM upregulated genes enriched inflammatory biological processes. Digital cytometry revealed that LM patients presented high proportions of cytotoxic and exhausted CD8+ T cells. CONCLUSIONS Our prediction model identified cutoffs that distinguished patients with lower PMA and survival with an inflammatory and immunosuppressive TME enriched with inflammatory factors and CD8+ T cells.
Collapse
Affiliation(s)
- Sarah Santiloni Cury
- grid.410543.70000 0001 2188 478XDepartment of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo CEP: 18.618-689 Brazil
| | - Diogo de Moraes
- grid.410543.70000 0001 2188 478XDepartment of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo CEP: 18.618-689 Brazil ,grid.411087.b0000 0001 0723 2494Department of Biochemistry and Tissue Biology, University of Campinas, Rua Monteiro Lobato, 255, Campinas, São Paulo 13083-862 Brazil
| | - Jakeline Santos Oliveira
- grid.410543.70000 0001 2188 478XDepartment of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo CEP: 18.618-689 Brazil
| | - Paula Paccielli Freire
- grid.11899.380000 0004 1937 0722Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP Brazil
| | - Patricia Pintor dos Reis
- grid.410543.70000 0001 2188 478XDepartment of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo 18618687 Brazil
| | - Miguel Luiz Batista
- grid.189504.10000 0004 1936 7558Department of Biochemistry, Boston University School of Medicine, Boston, USA
| | - Érica Nishida Hasimoto
- grid.410543.70000 0001 2188 478XDepartment of Surgery and Orthopedics, Faculty of Medicine, São Paulo State University (UNESP), Botucatu, São Paulo 18618687 Brazil
| | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, CEP: 18.618-689, Brazil.
| |
Collapse
|
11
|
Ferrara M, Samaden M, Ruggieri E, Vénéreau E. Cancer cachexia as a multiorgan failure: Reconstruction of the crime scene. Front Cell Dev Biol 2022; 10:960341. [PMID: 36158184 PMCID: PMC9493094 DOI: 10.3389/fcell.2022.960341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Cachexia is a devastating syndrome associated with the end-stage of several diseases, including cancer, and characterized by body weight loss and severe muscle and adipose tissue wasting. Although different cancer types are affected to diverse extents by cachexia, about 80% of all cancer patients experience this comorbidity, which highly reduces quality of life and response to therapy, and worsens prognosis, accounting for more than 25% of all cancer deaths. Cachexia represents an urgent medical need because, despite several molecular mechanisms have been identified, no effective therapy is currently available for this devastating syndrome. Most studies focus on skeletal muscle, which is indeed the main affected and clinically relevant organ, but cancer cachexia is characterized by a multiorgan failure. In this review, we focus on the current knowledge on the multiple tissues affected by cachexia and on the biomarkers with the attempt to define a chronological pathway, which might be useful for the early identification of patients who will undergo cachexia. Indeed, it is likely that the inefficiency of current therapies might be attributed, at least in part, to their administration in patients at the late stages of cachexia.
Collapse
Affiliation(s)
- Michele Ferrara
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Samaden
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Elena Ruggieri
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Emilie Vénéreau
- Tissue Regeneration and Homeostasis Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
12
|
Dhiman P, Ma J, Andaur Navarro CL, Speich B, Bullock G, Damen JAA, Hooft L, Kirtley S, Riley RD, Van Calster B, Moons KGM, Collins GS. Methodological conduct of prognostic prediction models developed using machine learning in oncology: a systematic review. BMC Med Res Methodol 2022; 22:101. [PMID: 35395724 PMCID: PMC8991704 DOI: 10.1186/s12874-022-01577-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/18/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Describe and evaluate the methodological conduct of prognostic prediction models developed using machine learning methods in oncology. METHODS We conducted a systematic review in MEDLINE and Embase between 01/01/2019 and 05/09/2019, for studies developing a prognostic prediction model using machine learning methods in oncology. We used the Transparent Reporting of a multivariable prediction model for Individual Prognosis Or Diagnosis (TRIPOD) statement, Prediction model Risk Of Bias ASsessment Tool (PROBAST) and CHecklist for critical Appraisal and data extraction for systematic Reviews of prediction Modelling Studies (CHARMS) to assess the methodological conduct of included publications. Results were summarised by modelling type: regression-, non-regression-based and ensemble machine learning models. RESULTS Sixty-two publications met inclusion criteria developing 152 models across all publications. Forty-two models were regression-based, 71 were non-regression-based and 39 were ensemble models. A median of 647 individuals (IQR: 203 to 4059) and 195 events (IQR: 38 to 1269) were used for model development, and 553 individuals (IQR: 69 to 3069) and 50 events (IQR: 17.5 to 326.5) for model validation. A higher number of events per predictor was used for developing regression-based models (median: 8, IQR: 7.1 to 23.5), compared to alternative machine learning (median: 3.4, IQR: 1.1 to 19.1) and ensemble models (median: 1.7, IQR: 1.1 to 6). Sample size was rarely justified (n = 5/62; 8%). Some or all continuous predictors were categorised before modelling in 24 studies (39%). 46% (n = 24/62) of models reporting predictor selection before modelling used univariable analyses, and common method across all modelling types. Ten out of 24 models for time-to-event outcomes accounted for censoring (42%). A split sample approach was the most popular method for internal validation (n = 25/62, 40%). Calibration was reported in 11 studies. Less than half of models were reported or made available. CONCLUSIONS The methodological conduct of machine learning based clinical prediction models is poor. Guidance is urgently needed, with increased awareness and education of minimum prediction modelling standards. Particular focus is needed on sample size estimation, development and validation analysis methods, and ensuring the model is available for independent validation, to improve quality of machine learning based clinical prediction models.
Collapse
Affiliation(s)
- Paula Dhiman
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Jie Ma
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Constanza L Andaur Navarro
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cochrane Netherlands, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Benjamin Speich
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Garrett Bullock
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Johanna A A Damen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cochrane Netherlands, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Lotty Hooft
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cochrane Netherlands, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Shona Kirtley
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Richard D Riley
- Centre for Prognosis Research, School of Medicine, Keele University, Staffordshire, ST5 5BG, UK
| | - Ben Van Calster
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
- EPI-centre, KU Leuven, Leuven, Belgium
| | - Karel G M Moons
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Cochrane Netherlands, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Gary S Collins
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7LD, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
13
|
Paval DR, Patton R, McDonald J, Skipworth RJE, Gallagher IJ, Laird BJ. A systematic review examining the relationship between cytokines and cachexia in incurable cancer. J Cachexia Sarcopenia Muscle 2022; 13:824-838. [PMID: 35080147 PMCID: PMC8977958 DOI: 10.1002/jcsm.12912] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer cachexia is an unmet clinical need that affects more than 50% of patients with cancer. The systemic inflammatory response, which is mediated by a network of cytokines, has an established role in the genesis and maintenance of cancer as well as in cachexia; yet, the specific role of the cytokine milieu in cachexia requires elucidation. This systematic review aims to examine the relationship between cytokines and the cachexia syndrome in patients with incurable cancer. The databases MEDLINE, EMBASE, CINAHL, CENTRAL, PsycINFO, and Web of Science were searched for studies published between 01/01/2004 and 06/01/2020. Included studies measured cytokines and their relationship with cachexia and related symptoms/signs in adults with incurable cancer. After title screening (n = 5202), the abstracts (n = 1264) and the full-text studies (n = 322) were reviewed independently by two authors. The quality assessment of the selected papers was conducted using the modified Downs and Black checklist. Overall, 1277 patients with incurable cancer and 155 healthy controls were analysed in the 17 eligible studies. The mean age of the patients was 64 ± 15 (mean ± standard deviation). Only 34% of included participants were female. The included studies were assessed as moderate-quality to high-quality evidence (mean quality score: 7.8; range: 5-10). A total of 31 cytokines were examined in this review, of which interleukin-6 (IL-6, 14 studies) and tumour necrosis factor-α (TNF-α, 12 studies) were the most common. The definitions of cachexia and the weight-loss thresholds were highly variable across studies. Although the data could not be meta-analysed due to the high degree of methodological heterogeneity, the findings were discussed in a systematic manner. IL-6, TNF-α, and IL-8 were greater in cachectic patients compared with healthy individuals. Also, IL-6 levels were higher in cachectic participants as opposed to non-cachectic patients. Leptin, interferon-γ, IL-1β, IL-10, adiponectin, and ghrelin did not demonstrate any significant difference between groups when individuals with cancer cachexia were compared against non-cachectic patients or healthy participants. These findings suggest that a network of cytokines, commonly IL-6, TNF-α, and IL-8, are associated with the development of cachexia. Yet, this relationship is not proven to be causative and future studies should opt for longitudinal designs with consistent methodological approaches, as well as adequate techniques for analysing and reporting the results.
Collapse
Affiliation(s)
- D Robert Paval
- Faculty of Health Sciences and Sport, University of Stirling, Stirling, UK
| | | | | | | | - Iain J Gallagher
- Faculty of Health Sciences and Sport, University of Stirling, Stirling, UK
| | - Barry J Laird
- St Columba's Hospice, Edinburgh, UK.,Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | | |
Collapse
|
14
|
Fram J, Vail C, Roy I. Assessment of Cancer-Associated Cachexia - How to Approach Physical Function Evaluation. Curr Oncol Rep 2022; 24:751-761. [PMID: 35305209 DOI: 10.1007/s11912-022-01258-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Cachexia is a devastating syndrome that impacts a majority of cancer patients. Early assessment of cachexia is critical to implementing cachexia treatments. Our aim was to summarize the existing cachexia assessment tools for their utility in both symptom and function evaluation. RECENT FINDINGS Several tools now exist that provide a symptom-based approach for evaluating weight change, appetite, and nutrition impact symptoms in cancer patients with cachexia. However, current instruments used to assess physical function changes related to cachexia are limited in depth and breadth. Instead, we recommend a tiered approach to cachexia-related functional assessment that involves evaluation of activities of daily living, general mobility, and exercise tolerance in a prioritized sequence. Current tools for cancer-associated cachexia assessment are adept at symptom evaluation. New approaches to physical function evaluation are needed that efficiently and broadly evaluate the diverse functional needs of cachexia patients.
Collapse
Affiliation(s)
- Julia Fram
- Shirley Ryan AbilityLab, 26th floor, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine & Rehabilitation, Northwestern University Feinberg School of Medicine, 710 N Lake Shore Dr #1022, Chicago, IL, 60611, USA
| | - Caroline Vail
- Shirley Ryan AbilityLab, 26th floor, 355 E. Erie St, Chicago, IL, 60611, USA
- Department of Physical Medicine & Rehabilitation, Northwestern University Feinberg School of Medicine, 710 N Lake Shore Dr #1022, Chicago, IL, 60611, USA
| | - Ishan Roy
- Shirley Ryan AbilityLab, 26th floor, 355 E. Erie St, Chicago, IL, 60611, USA.
- Department of Physical Medicine & Rehabilitation, Northwestern University Feinberg School of Medicine, 710 N Lake Shore Dr #1022, Chicago, IL, 60611, USA.
- Robert H. Lurie Cancer Center, 675 N St Clair St Fl 21 Ste 100, Chicago, IL, 60611, USA.
| |
Collapse
|
15
|
Martin L, Muscaritoli M, Bourdel-Marchasson I, Kubrak C, Laird B, Gagnon B, Chasen M, Gioulbasanis I, Wallengren O, Voss AC, Goldwasser F, Jagoe RT, Deans C, Bozzetti F, Strasser F, Thoresen L, Kazemi S, Baracos V, Senesse P. Diagnostic criteria for cancer cachexia: reduced food intake and inflammation predict weight loss and survival in an international, multi-cohort analysis. J Cachexia Sarcopenia Muscle 2021; 12:1189-1202. [PMID: 34448539 PMCID: PMC8517347 DOI: 10.1002/jcsm.12756] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/26/2021] [Accepted: 06/15/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Cancer-associated weight loss (WL) associates with increased mortality. International consensus suggests that WL is driven by a variable combination of reduced food intake and/or altered metabolism, the latter often represented by the inflammatory biomarker C-reactive protein (CRP). We aggregated data from Canadian and European research studies to evaluate the associations of reduced food intake and CRP with cancer-associated WL (primary endpoint) and overall survival (OS, secondary endpoint). METHODS The data set included a total of 12,253 patients at risk for cancer-associated WL. Patient-reported WL history (% in 6 months) and food intake (normal, moderately, or severely reduced) were measured in all patients; CRP (mg/L) and OS were measured in N = 4960 and N = 9952 patients, respectively. All measures were from a baseline assessment. Clinical variables potentially associated with WL and overall survival (OS) including age, sex, cancer diagnosis, disease stage, and performance status were evaluated using multinomial logistic regression MLR and Cox proportional hazards models, respectively. RESULTS Patients had a mean weight change of -7.3% (±7.1), which was categorized as: ±2.4% (stable weight; 30.4%), 2.5-5.9% (19.7%), 6.0-10.0% (23.2%), 11.0-14.9% (12.0%), ≥15.0% (14.6%). Normal food intake, moderately, and severely reduced food intake occurred in 37.9%, 42.8%, and 19.4%, respectively. In MLR, severe WL (≥15%) (vs. stable weight) was more likely (P < 0.0001) if food intake was moderately [OR 6.28, 95% confidence interval (CI 5.28-7.47)] or severely reduced [OR 18.98 (95% CI 15.30-23.56)]. In subset analysis, adjusted for food intake, CRP was independently associated (P < 0.0001) with ≥15% WL [CRP 10-100 mg/L: OR 2.00, (95% CI 1.58-2.53)] and [CRP > 100 mg/L: OR 2.30 (95% CI 1.62-3.26)]. Diagnosis, stage, and performance status, but not age or sex, were significantly associated with WL. Median OS was 9.9 months (95% CI 9.5-10.3), with median follow-up of 39.7 months (95% CI 38.8-40.6). Moderately and severely reduced food intake and CRP independently predicted OS (P < 0.0001). CONCLUSIONS Modelling WL as the dependent variable is an approach that can help to identify clinical features and biomarkers associated with WL. Here, we identify criterion values for food intake impairment and CRP that may improve the diagnosis and classification of cancer-associated cachexia.
Collapse
Affiliation(s)
- Lisa Martin
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | | | - Catherine Kubrak
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Barry Laird
- University of Edinburgh, European Palliative Care Research Center, Edinburgh, UK
| | - Bruno Gagnon
- Department of Family Medicine and Emergency Medicine, Université Laval, Laval, Quebec, Canada
| | - Martin Chasen
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Ioannis Gioulbasanis
- Department of Medical Oncology, Αnimus-Κyanous Stavros General Clinic - Larissa, Thessaly, Greece
| | - Ola Wallengren
- Clinical Nutrition Unit, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anne C Voss
- Global Research and Development (retired), Abbott Nutrition, Columbus, Ohio, USA
| | - Francois Goldwasser
- Medical Oncology, Cochin Hospital, APHP 5, University of Paris, Paris, France
| | - R Thomas Jagoe
- McGill Cancer Nutrition Rehabilitation Clinic, Jewish General Hospital, Montreal, Quebec, Canada
| | - Chris Deans
- Clinical and Surgical Sciences, School of Clinical Sciences and Community Health, University of Edinburgh, Royal Infirmary, Edinburgh, UK
| | | | - Florian Strasser
- Oncological Palliative Medicine, Division of Oncology, Department of Internal Medicine and Palliative Care Center, Cantonal Hospital, St. Gallen, Switzerland
| | - Lene Thoresen
- Oncology Clinic, St. Olavs University Hospital, Trondheim, Norway
| | - Sean Kazemi
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Vickie Baracos
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Pierre Senesse
- Clinical Nutrition and Gastroenterology Unit, Institut de recherche en Cancérologie de Montpellier (IRCM) Inserm U1194, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
16
|
Mu W, Katsoulakis E, Whelan CJ, Gage KL, Schabath MB, Gillies RJ. Radiomics predicts risk of cachexia in advanced NSCLC patients treated with immune checkpoint inhibitors. Br J Cancer 2021; 125:229-239. [PMID: 33828255 PMCID: PMC8292339 DOI: 10.1038/s41416-021-01375-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Approximately 50% of cancer patients eventually develop a syndrome of prolonged weight loss (cachexia), which may contribute to primary resistance to immune checkpoint inhibitors (ICI). This study utilised radiomics analysis of 18F-FDG-PET/CT images to predict risk of cachexia that can be subsequently associated with clinical outcomes among advanced non-small cell lung cancer (NSCLC) patients treated with ICI. METHODS Baseline (pre-therapy) PET/CT images and clinical data were retrospectively curated from 210 ICI-treated NSCLC patients from two institutions. A radiomics signature was developed to predict the cachexia with PET/CT images, which was further used to predict durable clinical benefit (DCB), progression-free survival (PFS) and overall survival (OS) following ICI. RESULTS The radiomics signature predicted risk of cachexia with areas under receiver operating characteristics curves (AUCs) ≥ 0.74 in the training, test, and external test cohorts. Further, the radiomics signature could identify patients with DCB from ICI with AUCs≥0.66 in these three cohorts. PFS and OS were significantly shorter among patients with higher radiomics-based cachexia probability in all three cohorts, especially among those potentially immunotherapy sensitive patients with PD-L1-positive status (p < 0.05). CONCLUSIONS PET/CT radiomics analysis has the potential to predict the probability of developing cachexia before the start of ICI, triggering aggressive monitoring to improve potential to achieve more clinical benefit.
Collapse
Affiliation(s)
- Wei Mu
- grid.468198.a0000 0000 9891 5233Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | | | - Christopher J. Whelan
- grid.468198.a0000 0000 9891 5233Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Kenneth L. Gage
- grid.468198.a0000 0000 9891 5233Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Matthew B. Schabath
- grid.468198.a0000 0000 9891 5233Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA ,grid.468198.a0000 0000 9891 5233Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Robert J. Gillies
- grid.468198.a0000 0000 9891 5233Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| |
Collapse
|
17
|
Wiegert EVM, de Oliveira LC, Calixto-Lima L, Chaves GV, Silva Lopes MS, Peres WAF. New cancer cachexia staging system for use in clinical practice. Nutrition 2021; 90:111271. [PMID: 34004417 DOI: 10.1016/j.nut.2021.111271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVES Previous attempts to classify cancer cachexia (CC) have demonstrated limitations regarding stages and diagnostic criteria. This study aims to develop and validate a new staging system for CC in patients with incurable cancer. METHODS This is an analysis of a database from a prospective cohort study of 1325 patients with advanced cancer referred for palliative care between 2016 and 2020. The cohort was randomly divided into two groups: Development (882 patients) and validation (443 patients) sets. A hierarchical cluster analysis was performed to distinguish different stages of CC in the development set. Next, the optimal cutoff points and ideal combinations of the most important factors associated with the CC groups (clusters) were ascertained. Finally, the relationship between the CC stages determined using the new system and body composition, quality of life, and overall survival was verified with the validation set. RESULTS The new system classified CC into three stages: Precachexia (10.8%), cachexia (57.8%), and refractory cachexia (31.4%), based on a combination of percentage weight loss in the past 6 mo (<15 or ≥15), body mass index (<21.0, 21.0-26.4, >26.4 kg/m2), and mid-upper-arm muscle area (≥38.0/≥35.5 or <38.0/<35.5 cm2 in men/women, respectively). The new staging system enabled a clear classification of patients into three CC groups according to the outcomes analyzed. Outcomes of patients with refractory cachexia were significantly worse than those in the other groups. CONCLUSIONS This study presents a useful, valid system for CC staging in the clinical setting, and is also capable of predicting outcomes, including quality of life and overall survival.
Collapse
Affiliation(s)
| | | | | | | | - Márcia Soares Silva Lopes
- Josué de Castro Institute of Nutrition, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
18
|
Nutritional Deficiencies in Radiotherapy-Treated Head and Neck Cancer Patients. J Clin Med 2021; 10:jcm10040574. [PMID: 33546506 PMCID: PMC7913750 DOI: 10.3390/jcm10040574] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 12/24/2022] Open
Abstract
Nutritional deficiencies (malnutrition, cachexia, sarcopenia, and unfavorable changes in the body composition) developing as a side effect of radiotherapy (RT) currently represents a significant but still inaccurately studied clinical problem in cancer patients. The incidence of malnutrition observed in head and neck cancer (HNC) patients in oncological radiology departments can reach 80%. The presence of malnutrition, sarcopenia, and cachexia is associated with an unfavorable prognosis of the disease, higher mortality, and deterioration of the quality of life. Therefore, it is necessary to identify patients with a high risk of both metabolic syndromes. However, the number of studies investigating potential predictive markers for the mentioned purposes is still significantly limited. This literature review summarizes the incidence of nutritional deficiencies in HNC patients prior to therapy and after the commencement of RT, and presents recent perspectives for the prediction of unfavorable nutritional changes developing as a result of applied RT.
Collapse
|
19
|
Masi T, Patel BM. Altered glucose metabolism and insulin resistance in cancer-induced cachexia: a sweet poison. Pharmacol Rep 2020; 73:17-30. [PMID: 33141425 DOI: 10.1007/s43440-020-00179-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022]
Abstract
Cancer cachexia is a wasting disorder characterised by specific skeletal muscle and adipose tissue loss. Cancer cachexia is also driven by inflammation, altered metabolic changes such as increased energy expenditure, elevated plasma glucose, insulin resistance and excess catabolism. In cachexia, host-tumor interaction causes release of the lactate and inflammatory cytokines. Lactate released by tumor cells takes part in hepatic glucose production with the help of gluconeogenic enzymes. Thus, Cori cycle between organs and cancerous cells contributes to increased glucose production and energy expenditure. A high amount of blood glucose leads to increased production of insulin. Overproduction of insulin causes inactivation of PI3K/Akt/m-TOR pathway and finally results in insulin resistance. Insulin is involved in maintaining the vitality of organs and regulate the metabolism of glucose, protein and lipids. Insulin insensitivity decreases the uptake of glucose in the organs and results in loss of skeletal muscles and adipose tissues. However, looking into the complexity of this metabolic syndrome, it is impossible to rely on a single variable to treat patients having cancer cachexia. Hence, it becomes greater a challenge to produce a clinically effective treatment for this metabolic syndrome. Thus, the present paper aims to provide an understanding of pathogenesis and mechanism underlining the altered glucose metabolism and insulin resistance and its contribution to the progression of skeletal muscle wasting and lipolysis, providing future direction of research to develop new pharmacological treatment in cancer cachexia.
Collapse
Affiliation(s)
- Tamhida Masi
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | - Bhoomika M Patel
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
20
|
Rosa-Caldwell ME, Fix DK, Washington TA, Greene NP. Muscle alterations in the development and progression of cancer-induced muscle atrophy: a review. J Appl Physiol (1985) 2019; 128:25-41. [PMID: 31725360 DOI: 10.1152/japplphysiol.00622.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Cancer cachexia-cancer-associated body weight and muscle loss-is a significant predictor of mortality and morbidity in cancer patients across a variety of cancer types. However, despite the negative prognosis associated with cachexia onset, there are no clinical therapies approved to treat or prevent cachexia. This lack of treatment may be partially due to the relative dearth of literature on mechanisms occurring within the muscle before the onset of muscle wasting. Therefore, the purpose of this review is to compile the current scientific literature on mechanisms contributing to the development and progression of cancer cachexia, including protein turnover, inflammatory signaling, and mitochondrial dysfunction. We define "development" as changes in cell function occurring before the onset of cachexia and "progression" as alterations to cell function that coincide with the exacerbation of muscle wasting. Overall, the current literature suggests that multiple aspects of cellular function, such as protein turnover, inflammatory signaling, and mitochondrial quality, are altered before the onset of muscle loss during cancer cachexia and clearly highlights the need to study more thoroughly the developmental stages of cachexia. The studying of these early aberrations will allow for the development of effective therapeutics to prevent the onset of cachexia and improve health outcomes in cancer patients.
Collapse
Affiliation(s)
- Megan E Rosa-Caldwell
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Dennis K Fix
- Molecular Medicine Program, University of Utah, Salt Lake City, Utah
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| | - Nicholas P Greene
- Integrative Muscle Metabolism Laboratory, Exercise Science Research Center, Department of Human Health Performance and Recreation, University of Arkansas, Fayetteville, Arkansas
| |
Collapse
|