1
|
Abdilleh K, Aguilar B, Acquaah-Mensah G. Clinical and Multiomic Features Differentiate Young Black and White Breast Cancer Cohorts Derived by Machine Learning Approaches. Clin Breast Cancer 2025; 25:e301-e311. [PMID: 39706709 PMCID: PMC11911081 DOI: 10.1016/j.clbc.2024.11.015] [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: 05/30/2024] [Revised: 09/30/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND There are documented differences in Breast cancer (BrCA) presentations and outcomes between Black and White patients. In addition to molecular factors, socioeconomic, racial, and clinical factors result in disparities in outcomes for women in the United States. Using machine learning and unsupervised biclustering methods within a multiomics framework, here we sought to shed light on the biological and clinical underpinnings of observed differences between Black and White BrCA patients. MATERIALS AND METHODS We examined The Cancer Genome Atlas BrCA samples from stage II patients aged 50 or younger that are Black (BAA50) or White (W50) (n = 139 patients; 36 BAA50 and 103 W50) These patients were chosen because marked differences in survival were observed in an earlier study. A variety of multiomic data sets were analyzed to further characterize the clinical and molecular disparities for insights. RESULTS We coupled RNAseq data with protein-protein interaction as well as BrCA-specific protein co-expression network data to identify 2 novel biclusters. These biclusters are significantly associated with clinical features including race, number of lymph nodes involved with disease, estrogen receptor status, progesterone receptor status and menopausal status. There were also differentially mutated genes. Using DNA methylation data, we identified differentially methylated genes. Machine learning algorithms were trained on differential methylation values of driver genes. The trained algorithms were successful in predicting the bicluster assignment of each sample. CONCLUSION These results demonstrate that there was a significant association between the cluster membership and BAA50 and W50 cohorts, indicating that these biclusters accurately stratify these cohorts.
Collapse
|
2
|
Bokemeyer F, Lebherz L, Bokemeyer C, Derksen JWG, Schulz H, Bleich C. Practice patterns, experiences, and challenges of German oncology health care staff with smoking cessation in patients with cancer: a cross-sectional survey study. J Cancer Surviv 2025; 19:701-712. [PMID: 38012516 PMCID: PMC11926055 DOI: 10.1007/s11764-023-01501-2] [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: 08/15/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE Often, cancer patients do not receive education about the negative consequences of smoking on the treatment outcome. To support cancer patients in the process of smoking cessation, it is essential to involve oncology staff. This study aims to learn about the experiences and attitudes from the point of view of oncology staff and, thus, how a smoking intervention should be designed. The study aims to engage all oncology staff due to the unclear responsibility for providing smoking cessation education, support, and motivating cancer patients to quit smoking. METHODS N = 354 German oncology staff (oncologists, nurses, psycho-oncologists, others) filled out a 5-point Likert scale-based questionnaire regarding practices, potential barriers, and attitudes towards smoking cessation between October 2021 and June 2022. The questionnaire was developed by Derksen et al. (2020), translated and slightly modified for the use of this study. It was distributed to all leading oncology staff in our Cancer Center Network with a request to share with all oncology staff. Flyers were also handed out in all oncology wards and outpatient clinics in the same Cancer Center Network. RESULTS Most oncology staff ask cancer patients about their current smoking status (curative, M = 2.27; SD = 1.59; palliative, M = 2.90; SD = 1.83), but they rarely treat or refer patients for a smoking cessation intervention (curative, M = 4.78; SD = 1.20; palliative, M = 4.99; SD = 1.06). Smoking behavior of curative cancer patients is addressed more than that of palliative cancer patients (d = - 37). Regression analyses of key dependent variables showed that profession, setting, and the belief that continued smoking affects treatment outcome explained the variance of asking patients if they smoke, advising to stop smoking and lack of time (without profession). CONCLUSION Involving oncology staff in motivating cancer patients who smoke to quit and referring them to smoking cessation services should take the different attitudes and knowledge of the staff into account to improve treatment that supports tobacco cessation. IMPLICATIONS FOR CANCER SURVIVORS Cancer patients have special needs when it comes to a cessation program. In the long term, survivors will benefit from tailored smoking cessation education and services provided by oncology staff to help them quit smoking after a cancer diagnosis.
Collapse
Affiliation(s)
- Frederike Bokemeyer
- Department of Medical Psychology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
- Center for Oncology, II. Medical Clinic and Polyclinic, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Lisa Lebherz
- Department of Medical Psychology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Carsten Bokemeyer
- Center for Oncology, II. Medical Clinic and Polyclinic, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Jeroen W G Derksen
- Division Julius Center for Health Sciences and Primary Care, Department of Epidemiology and Health Economics, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584, CX, Utrecht, The Netherlands
| | - Holger Schulz
- Department of Medical Psychology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Christiane Bleich
- Department of Medical Psychology, University Medical Center Hamburg Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
3
|
Nemade H, Thaduri A, Gondi JT, Chava S, Kumar A, Raj P, Neelap U, Akalankam P, Rukmangatham TM, Sekara Rao S LMC. Five-year long-term functional and quality of life outcomes in total glossectomy survivors. Eur Arch Otorhinolaryngol 2025; 282:2063-2070. [PMID: 39511055 DOI: 10.1007/s00405-024-09059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Total glossectomy is proven to be a superior treatment option for advanced tongue cancer. Despite its efficacy, the procedure is associated with profound morbidity, notably impacting Speech, swallowing, and overall quality of life. Limited therapeutic benefits and the potential for considerable morbidity render total glossectomy a subject of controversy. METHODS We performed a multidimensional quality of life evaluation of long-term (more than 5 yrs.) total glossectomy survivors. In this study, we evaluated 25 total glossectomy survivors with comprehensive functional and quality of life outcomes using objective measures (London Speech Evaluation Scale, FOIS, 100 ml Water Challenge, FEES) and Patient-reported outcomes (SHI, PSS HN, EORTC QLQ 30 & HN35). RESULTS EORTC QLQ 30 showed overall good global health status (M = 80 ± 20) and functional scale mean scores ranging from 87 to 91, despite issues in pain (M = 31.6 ± 31), swallowing (M = 23.3 ± 24), and Speech (26.1 ± 1). Speech outcomes revealed 80% with moderate to severe intelligibility impairment. Swallowing outcomes showed 84% requiring special food preparation, 24% exhibiting aspiration, and 72% having pharyngeal residue. Speech outcomes showed 88% having moderate-severe speech impairment, and Patient-reported speech outcomes indicated a mean SHI score of (M = 47.2 ± 42). CONCLUSION Patients undergoing total glossectomy report favourable long-term outcomes in global health-related quality of life despite moderate to severe impairments in objective speech and swallowing evaluations. These positive patient-reported outcomes support the consideration of total glossectomy as a viable treatment option if and when required, underscoring the importance of evaluating treatment outcomes through the patient-perceived quality of life.
Collapse
Affiliation(s)
- Hemant Nemade
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Abhinav Thaduri
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India.
| | - Jonathan T Gondi
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Sravankumar Chava
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Anil Kumar
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Pratheek Raj
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Uma Neelap
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | - Pardhasaradhi Akalankam
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| | | | - L M Chandra Sekara Rao S
- Head and Neck Services, Department of Surgical Oncology, Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, India
| |
Collapse
|
4
|
Chu YL, Georgeson P, Clendenning M, Mahmood K, Walker R, Como J, Joseland S, Preston SG, Rice T, Lynch BM, Milne RL, Southey MC, Giles GG, Phipps AI, Hopper JL, Win AK, Rosty C, Macrae FA, Winship I, Jenkins MA, Buchanan DD, Joo JE. Intratumoural pks +Escherichia coli is associated with risk of metachronous colorectal cancer and adenoma development in people with Lynch syndrome. EBioMedicine 2025; 114:105661. [PMID: 40158390 DOI: 10.1016/j.ebiom.2025.105661] [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: 10/23/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The adverse gut microbiome may underlie the variability in risks of colorectal cancer (CRC) and metachronous CRC in people with Lynch syndrome (LS). The role of pks+/-Escherichia coli (pks+/-E. coli), Enterotoxigenic Bacteroides fragilis (ETBF), and Fusobacterium nucleatum (Fn) in CRCs and adenomas in people with LS is unknown. METHODS A total of 358 LS cases, including 386 CRCs, 90 adenomas, 195 normal colonic mucosa DNA from the Australasian Colon Cancer Family Registry were tested using multiplex TaqMan qPCR. Logistic regression was used to compare the intratumoural prevalence of each bacteria in Lynch CRCs with 1336 sporadic CRCs. Cox proportional-hazards regression estimated the associations of each bacteria with the risk of metachronous CRC and neoplasia. FINDINGS Pks+ E. coli (odds ratio [95% confidence interval] = 1.60 [1.08-2.35], P = 0.017), pks-E. coli (3.87 [2.58-5.80], P < 0.001) and Fn (19.47 [13.32-28.87], P < 0.001) were significantly enriched in LS CRCs when compared with sporadic CRCs. Pks+ E. coli in the initial CRC was associated with an increased risk of metachronous CRC (hazard ratio [95% confidence interval] = 2.32 [1.29-4.17], P = 0.005) and metachronous colorectal neoplasia (1.51 [1.02-2.23], P = 0.040) when compared with CRCs without pks+ E. coli. INTERPRETATION Pks+ E. coli, pks-E. coli, and Fn are enriched within LS CRCs, suggesting possible roles in CRC development in LS. Having intratumoural pks+ E. coli is associated with increased risk of metachronous CRC, suggesting that, if validated, people with LS might benefit from pks+ E. coli screening and eradication. FUNDING This work was funded by an NHMRC Investigator grant (GNT1194896) and a Cancer Australia/Cancer Council NSW co-funded grant (GNT2012914).
Collapse
Affiliation(s)
- Yen Lin Chu
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Clendenning
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Khalid Mahmood
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Melbourne Bioinformatics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Romy Walker
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Julia Como
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Sharelle Joseland
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Susan G Preston
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Toni Rice
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia
| | - Brigid M Lynch
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Melissa C Southey
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria, Australia
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Amanda I Phipps
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Aung K Win
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Christophe Rosty
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; University of Queensland, Brisbane, Queensland, Australia; Envoi Specialist Pathologists, Brisbane, Queensland, Australia
| | - Finlay A Macrae
- Colorectal Medicine and Genetics, The Royal Melbourne Hospital, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Ingrid Winship
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Melbourne, Victoria, Australia
| | - Jihoon E Joo
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia; Collaborative Centre for Genomic Cancer Medicine, Victorian Comprehensive Cancer Centre, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
5
|
Holthuijsen DDB, van Roekel EH, Bours MJL, Ueland PM, Breukink SO, Janssen-Heijnen MLG, Konsten JL, Keulen ETP, McCann A, Brezina S, Gigic B, Kok DE, Ulrich CM, Weijenberg MP, Eussen SJPM. Modeling how iso-caloric macronutrient substitutions are longitudinally associated with plasma kynurenines in colorectal cancer survivors up to 12 months posttreatment. J Nutr Biochem 2025:109910. [PMID: 40158742 DOI: 10.1016/j.jnutbio.2025.109910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/08/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
Dietary intake of several macronutrients is associated with plasma kynurenines after colorectal cancer (CRC), and kynurenines have been linked to health-related outcomes. It is unknown how macronutrient substitution affects plasma kynurenines, which may be relevant for developing guidelines to improve post-CRC quality of life through dietary changes. Using iso-caloric substitution models, we investigated how substituting one macronutrient with another is longitudinally associated with plasma tryptophan, kynurenines, and kynurenine ratios in CRC survivors. Measurements were performed at 6-weeks, 6-months, and 12-months posttreatment in 247 stage I-III CRC survivors. Macronutrient intake was measured by seven-day dietary records and plasma kynurenines by LC/MS-MS. For analysis, we applied linear mixed models with false discovery rate (FDR) to adjust for multiple testing. After FDR adjustment, substituting 100 kcal/day of total carbohydrates with 100 kcal/day of total protein was associated with higher plasma concentrations of kynurenic acid (KA), xanthurenic acid (XA), and a higher kynurenic acid-to-quinolinic acid (KA/QA) ratio. Substituting 100 kcal/day of total carbohydrates with 100 kcal/day of total fat was associated with higher tryptophan concentrations, higher KA/QA ratio, and a lower kynurenine-to-tryptophan ratio (KTR) and hydroxykynurenine ratio (HKr). Substituting 100 kcal/day of total fat with 100 kcal/day of total protein was associated with higher XA concentrations. Altogether, iso-caloric macronutrient substitutions, particularly substituting carbohydrates with protein or fat, were longitudinally associated with higher concentrations of potentially favourable kynurenines and ratios (i.e., KA, XA, and KA/QA ratio) and lower ratios with pro-inflammatory or neurotoxic properties (i.e., KTR and HKr) in CRC survivors up to 12-months posttreatment.
Collapse
Affiliation(s)
- Daniëlle D B Holthuijsen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands.
| | - Eline H van Roekel
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Martijn J L Bours
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | | | - Stéphanie O Breukink
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands; NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Maryska L G Janssen-Heijnen
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Clinical Epidemiology, VieCuri Medical Centre, Venlo, The Netherlands
| | - Joop L Konsten
- Department of Surgery, VieCuri Medical Centre, Venlo, The Netherlands
| | - Eric T P Keulen
- Department of Internal Medicine and Gastroenterology, Zuyderland Medical Centre Sittard-Geleen, Geleen, The Netherlands
| | | | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Dieuwertje E Kok
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Matty P Weijenberg
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands; Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
6
|
Rahadi A, Mahardya RTK, Listiani P, Herlinawaty E, Nugraha RR, Budiman DR, Suharlim C. Calibration of transition probabilities to model survival of adjuvant trastuzumab for early breast cancer in Indonesia. Int J Technol Assess Health Care 2025; 41:e18. [PMID: 40135279 PMCID: PMC11955306 DOI: 10.1017/s0266462325000157] [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: 07/21/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVES Cost-effectiveness models fully informed by real-world epidemiological parameters yield the best results, but they are costly to obtain. Model calibration using real-world data/evidence (RWD/E) on routine health indicators can provide an alternative to improve the validity and acceptability of the results. We calibrated the transition probabilities of the reference chemotherapy treatment using RWE on patient overall survival (OS) to model the survival benefit of adjuvant trastuzumab in Indonesia. METHODS A Markov model comprising four health states was initially parameterized using the reference-treatment transition probabilities, obtained from published international evidence. We then calibrated these probabilities, targeting a 2-year OS of 86.11 percent from the RWE sourced from hospital registries. We compared projected OS duration and life-years gained (LYG) before and after calibration for the Nelder-Mead, Bound Optimization BY Quadratic Approximation, and generalized reduced gradient (GRG) nonlinear optimization methods. RESULTS The pre-calibrated transition probabilities overestimated the 2-year OS (92.25 percent). GRG nonlinear performed best and had the smallest difference with the RWD/E OS. After calibration, the projected OS duration was significantly lower than their pre-calibrated estimates across all optimization methods for both standard chemotherapy (~7.50 vs. 11.00 years) and adjuvant trastuzumab (~9.50 vs. 12.94 years). LYG measures were, however, similar (~2 years) for the pre-calibrated and calibrated models. CONCLUSIONS RWD/E calibration resulted in realistically lower survival estimates. Despite the little difference in LYG, calibration is useful to adapt external evidence commonly used to derive transition probabilities to the policy context, thereby enhancing the validity and acceptability of the modeling results.
Collapse
Affiliation(s)
- Arie Rahadi
- Management Sciences for Health, Arlington, VA, USA
| | | | - Putri Listiani
- Center for Health Financing Policy and Insurance Management, Gadjah Mada University, Sleman, Yogyakarta, Indonesia
| | - Eva Herlinawaty
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | | - Dani Ramdhani Budiman
- Center for Health Financing and Decentralization Policy, Ministry of Health Republic of Indonesia, Central Jakarta, Jakarta, Indonesia
| | | |
Collapse
|
7
|
Fortunato A, Mallo D, Cisneros L, King LM, Khan A, Curtis C, Ryser MD, Lo JY, Hall A, Marks JR, Hwang ES, Maley CC. Evolutionary measures show that recurrence of DCIS is distinct from progression to breast cancer. Breast Cancer Res 2025; 27:43. [PMID: 40119428 PMCID: PMC11929273 DOI: 10.1186/s13058-025-01966-2] [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: 09/04/2024] [Accepted: 01/19/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND Progression from pre-cancers like ductal carcinoma in situ (DCIS) to invasive disease (cancer) is driven by somatic evolution and is altered by clinical interventions. We hypothesized that genetic and/or phenotypic intra-tumor heterogeneity would predict clinical outcomes for DCIS since it serves as the substrate for natural selection among cells. METHODS We profiled two samples from two geographically distinct foci from each DCIS in both cross-sectional (n = 119) and longitudinal cohorts (n = 224), with whole exome sequencing, low-pass whole genome sequencing, and a panel of immunohistochemical markers. RESULTS In the longitudinal cohorts, the only statistically significant associations with time to non-invasive DCIS recurrence were the combination of treatment (lumpectomy only vs mastectomy or lumpectomy with radiation, HR 12.13, p = 0.003, Wald test with FDR correction), ER status (HR 0.16 for ER+ compared to ER-, p = 0.0045), and divergence in SNVs between the two samples (HR 1.33 per 10% divergence, p = 0.018). SNV divergence also distinguished between pure DCIS and DCIS synchronous with invasive disease in the cross-sectional cohort. In contrast, the only statistically significant associations with time to progression to invasive disease were the combination of the width of the surgical margin (HR 0.67 per mm, p = 0.043) and the number of mutations that were detectable at high allele frequencies (HR 1.30 per 10 SNVs, p = 0.02). No predictors were significantly associated with both DCIS recurrence and progression to invasive disease, suggesting that the evolutionary scenarios that lead to these clinical outcomes are markedly different. CONCLUSIONS These results imply that recurrence with DCIS is a clinical and biological process different from invasive progression.
Collapse
Affiliation(s)
- Angelo Fortunato
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ, 85281, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ, 85287, USA
| | - Diego Mallo
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ, 85281, USA
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ, 85287, USA
| | - Luis Cisneros
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ, 85281, USA
- Mayo Clinic OPART Oncology Department, Mayo Clinic, Rochester, MN, USA
| | | | - Aziz Khan
- Department of Medicine, Genetics, and Biomedical Data Science Stanford School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Christina Curtis
- Department of Medicine, Genetics, and Biomedical Data Science Stanford School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford School of Medicine, Stanford, CA, 94305, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Marc D Ryser
- Duke University School of Medicine, Durham, NC, 27710, USA
| | - Joseph Y Lo
- Duke University School of Medicine, Durham, NC, 27710, USA
| | - Allison Hall
- Duke University School of Medicine, Durham, NC, 27710, USA
| | | | | | - Carlo C Maley
- Arizona Cancer Evolution Center and Biodesign Center for Biocomputing, Security and Society, Arizona State University, 727 E. Tyler St., Tempe, AZ, 85281, USA.
- School of Life Sciences, Arizona State University, 427 East Tyler Mall, Tempe, AZ, 85287, USA.
| |
Collapse
|
8
|
Damiano OM, Stevens AJ, Kenwright DN, Seddon AR. Chronic Inflammation to Cancer: The Impact of Oxidative Stress on DNA Methylation. FRONT BIOSCI-LANDMRK 2025; 30:26142. [PMID: 40152377 DOI: 10.31083/fbl26142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 03/29/2025]
Abstract
The genomic landscape of cancer cells is complex and heterogeneous, with aberrant DNA methylation being a common observation. Growing evidence indicates that oxidants produced from immune cells may interact with epigenetic processes, and this may represent a mechanism for the initiation of altered epigenetic patterns observed in both precancerous and cancerous cells. Around 20% of cancers are linked to chronic inflammatory conditions, yet the precise mechanisms connecting inflammation with cancer progression remain unclear. During chronic inflammation, immune cells release oxidants in response to stimuli, which, in high concentrations, can cause cytotoxic effects. Oxidants are known to damage DNA and proteins and disrupt normal signalling pathways, potentially initiating a sequence of events that drives carcinogenesis. While research on the impact of immune cell-derived oxidants on DNA methylation remains limited, this mechanism may represent a crucial link between chronic inflammation and cancer development. This review examines current evidence on inflammation-associated DNA methylation changes in cancers related to chronic inflammation.
Collapse
Affiliation(s)
- Olivia M Damiano
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Aaron J Stevens
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Diane N Kenwright
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
| | - Annika R Seddon
- Genetics and Epigenetics Research Group, Department of Pathology and Molecular Medicine, University of Otago, 6021 Wellington, New Zealand
- Mātai Hāora - Centre for Redox Biology and Medicine, Department of Pathology and Biomedical Science, University of Otago, 8011 Christchurch, New Zealand
| |
Collapse
|
9
|
Dardenne A, Dhooge M, Basset N, Chansavang A, Metras J, Farelly S, Netter J, Coulet F, Benusiglio PR. Pathogenic germline variants in patients with early-onset colorectal cancer according to phenotype. Eur J Hum Genet 2025:10.1038/s41431-025-01808-x. [PMID: 40097831 DOI: 10.1038/s41431-025-01808-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 01/22/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
We assessed retrospectively the prevalence of pathogenic germline variants (PGV) in 268 French adult patients diagnosed with colorectal cancer (CRC) before age 41, stratified by phenotype. APC, BMPR1A, CDH1, EPCAM, MLH1, MSH2, MSH3, MSH6, MUTYH, NTHL1, POLE, POLD1, PTEN, PMS2, SMAD4, STK11 and TP53 were analyzed. Overall, 21.6% of cases carried a PGV. A high prevalence was observed in Mismatch Repair-deficient (MMRd) CRC (60.1%, MMR genes) and polyposis-associated CRC (48%, APC, MUTYH and MSH3-biallelic, POLE). Only 2.3% of patients with MMR proficient and without polyposis carried a PGV. The genes involved in this third group were POLE and MSH2, and three out of four cases had either two synchronous CRC or a CRC family history. Phenotypic features should be taken into account for testing decision. Evaluating the cost-effectiveness of testing all CRC cases < 41 years, as well as how it aligns with the constraints of various healthcare systems, is warranted.
Collapse
Affiliation(s)
- Antoine Dardenne
- Sorbonne Université, Service de Chirurgie digestive, Hôpital Saint-Antoine, APHP, Paris, France
| | - Marion Dhooge
- Paris Cité Université, Service de Gastroentérologie et Oncologie digestive, UF d'Oncogénétique digestive, Hôpital Cochin, APHP, Paris, France
| | - Noémie Basset
- Sorbonne Université, Hôpital Pitié-Salpêtrière, Département de Génétique médicale, APHP, Paris, France
| | - Albain Chansavang
- Département de Médecine Génomique des Tumeurs et Cancers, Hôpital Cochin, APHP, Université Paris Cité, Paris, France
| | - Julie Metras
- Sorbonne Université, Service de Chirurgie digestive, Hôpital Saint-Antoine, APHP, Paris, France
| | - Solenne Farelly
- Paris Cité Université, Service de Gastroentérologie et Oncologie digestive, UF d'Oncogénétique digestive, Hôpital Cochin, APHP, Paris, France
| | - Jeanne Netter
- Sorbonne Université, Service de Chirurgie digestive, Hôpital Saint-Antoine, APHP, Paris, France
- Service d'Oncologie digestive, Hôpital Européen Georges Pompidou, APHP, Université Paris Cité, Paris, France
| | - Florence Coulet
- Sorbonne Université, Hôpital Pitié-Salpêtrière, Département de Génétique médicale, APHP, Paris, France
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 et SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Paris, France
| | - Patrick R Benusiglio
- Sorbonne Université, Service de Chirurgie digestive, Hôpital Saint-Antoine, APHP, Paris, France.
- Sorbonne Université, Hôpital Pitié-Salpêtrière, Département de Génétique médicale, APHP, Paris, France.
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 et SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Paris, France.
| |
Collapse
|
10
|
Ji X, Hao M, Wang Y, Kong W, Pan Z, Sun Q, Miao J. Human papillomavirus self-sampling in Asia: a systematic review. Front Microbiol 2025; 16:1540609. [PMID: 40160269 PMCID: PMC11949917 DOI: 10.3389/fmicb.2025.1540609] [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: 12/06/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Background Human papillomavirus (HPV) self-sampling may be an accurate and effective alternative sampling method to conventional cervical cancer screening methods. This systematic review compares the accuracy and acceptance of self-sampling to clinician sampling for HPV testing in Asia. Methods The PubMed, Cochrane Library, Cumulative Index to Nursing and Allied Health, and Web of Science databases were searched for publications published from the establishment of the database to 2023. The risk of bias was assessed using the QUADAS-2 tool for studies included in this review. All studies evaluating the accuracy and acceptance of HPV self-sampling, and agreement of self- and clinician-collected samples in Asia were included. The accuracy of each study was demonstrated through the sensitivity and specificity in diagnosing cervical intraepithelial neoplasia or cancer, as well as the detection rate of HPV. The agreement between the two sampling methods was assessed based on the detection outcomes of HPV. Acceptance was indicated by women's preferences for HPV self-sampling. Results Sixty-seven studies including 117,279 adult, female participants were included in this review. The type of HPV screening, other intervention components, study design, sample size, follow-up period, analysis method, numerical outcomes, results, and limitations were extracted from each study. The sensitivity and specificity of HPV self-sampling in detecting cervical intraepithelial neoplasia were higher than 80% and 70%, consistent with the results of HPV clinician sampling. The consistency between self-sampling and clinician-sampling was high in most studies, and the kappa value was more than 0.7. Women had high acceptance of self-sampling but expressed some concerns. Conclusion Self-sampling for HPV testing can significantly improve cervical cancer screening coverage, especially in areas with limited medical resources or reluctance to accept physician sampling. In most studies, the accuracy and acceptance of HPV self-sampling was comparable to clinician sampling. However, the diagnostic criteria and HPV detection methods still need to be adjusted due to the low sensitivity of HPV self-sampling in some studies in China and India. Targeted health education should be carried out to improve the acceptance of HPV self-sampling in women. Systematic review registration https://inplasy.com/?s=INPLASY202520107, INPLASY202520107.
Collapse
Affiliation(s)
- Xuechao Ji
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Menglin Hao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Yixiao Wang
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Wenzhi Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Zangyu Pan
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Qi Sun
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
| | - Jinwei Miao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Beijing Maternal and Child Health Care Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
11
|
Ashtree DN, Orr R, Lane MM, Akbaraly TN, Bonaccio M, Costanzo S, Gialluisi A, Grosso G, Lassale C, Martini D, Monasta L, Santomauro D, Stanaway J, Jacka FN, O'Neil A. Estimating the Burden of Common Mental Disorders Attributable to Lifestyle Factors: Protocol for the Global Burden of Disease Lifestyle and Mental Disorder (GLAD) Project. JMIR Res Protoc 2025; 14:e65576. [PMID: 40085831 PMCID: PMC11953606 DOI: 10.2196/65576] [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: 08/19/2024] [Revised: 12/06/2024] [Accepted: 12/26/2024] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) collects and calculates risk-outcome data for modifiable lifestyle exposures (eg, dietary intake) and physical health outcomes (eg, cancers). These estimates form a critical digital resource tool, the GBD VizHub data visualization tool, for governments and policy makers to guide local, regional, and global health decisions. Despite evidence showing the contributions of lifestyle exposures to common mental disorders (CMDs), such as depression and anxiety, GBD does not currently generate these lifestyle exposure-mental disorder outcome pairings. This gap is due to a lack of uniformly collected and analyzed data about these exposures as they relate to CMDs. Such data are required to quantify whether, and to what degree, the global burden of CMDs could be reduced by targeting lifestyle factors at regional and global levels. We have established the Global burden of disease Lifestyle And mental Disorder (GLAD) Taskforce to address this gap. OBJECTIVE This study aims to generate the necessary estimates to afford the inclusion of lifestyle exposures as risk factors for CMDs in the GBD study and the GBD digital visualization tools, initially focusing on the relationship between dietary intake and CMDs. METHODS The GLAD project is a multicenter, collaborative effort to integrate lifestyle exposures as risk factors for CMDs in the GBD study. To achieve this aim, global epidemiological studies will be recruited to conduct harmonized data analyses estimating the risk, odds, or hazards of lifestyle exposures with CMD outcomes. Initially, these models will focus on the relationship between dietary intake, as defined by the GBD, and anxiety and depression. RESULTS As of August 2024, 18 longitudinal cohort studies from 9 countries (Australia: n=4; Brazil: n=1; France: n=1; Italy: n=3; The Netherlands: n=3; New Zealand: n=1; South Africa: n=1; Spain: n=1; and United Kingdom: n=3) have agreed to participate in the GLAD project. CONCLUSIONS Our comprehensive, collaborative approach allows for the concurrent execution of a harmonized statistical analysis protocol across multiple, internationally renowned epidemiological cohorts. These results will be used to inform the GBD study and incorporate lifestyle risk factors for CMD in the GBD digital platform. Consequently, given the worldwide influence of the GBD study, findings from the GLAD project can offer valuable insights to policy makers worldwide around lifestyle-based mental health care. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/65576.
Collapse
Affiliation(s)
- Deborah N Ashtree
- IMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Rebecca Orr
- IMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Melissa M Lane
- IMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| | - Tasnime N Akbaraly
- Université Montpellier, Institut National de Santé et de Recherche Médicale (INSERM), Desbrest Institute of Epidemiology and Public Health (IDESP), F-34090 Montpellier, France
| | - Marialaura Bonaccio
- IRCCS Neuromed, Research Unit of Epidemiology and Prevention, Pozzilli, Italy
| | - Simona Costanzo
- IRCCS Neuromed, Research Unit of Epidemiology and Prevention, Pozzilli, Italy
| | - Alessandro Gialluisi
- IRCCS Neuromed, Research Unit of Epidemiology and Prevention, Pozzilli, Italy
- Department of Medicine and Surgery, Libera Università Mediterranea (LUM) University, Casamassima (Bari), Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Camille Lassale
- ISGlobal, Barcelona, Spain
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBEROBN), Madrid, Spain
| | - Daniela Martini
- Division of Human Nutrition, Environmental and Nutritional Sciences, University of Milan, DeFENS-Department of Food, Milan, Italy
| | - Lorenzo Monasta
- Institute for Maternal and Child Health - IRCCS Burlo Garofolo, Trieste, Italy
| | - Damian Santomauro
- Queensland Centre for Mental Health Research, Wacol, Australia
- Faculty of Medicine, School of Public Health, University of Queensland, Herston, Australia
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, United States
| | - Jeffrey Stanaway
- Institute for Health Metrics and Evaluation, University of Washington, Seattle, WA, United States
| | - Felice N Jacka
- IMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, Australia
- Department of Immunology, Therapeutics, and Vaccines, James Cook University, Queensland, Australia
| | - Adrienne O'Neil
- IMPACT (the Institute for Mental and Physical Health and Clinical Translation), Food & Mood Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Australia
| |
Collapse
|
12
|
Aronson M, Palma L, Semotiuk K, Nuk J, Pollett A, Singh H, Rothenmund H, Racher H, Jessen J, Pautler SE, Rusnak A, Rutka M, Etchegary H, Tiano T, Kaurah P, Dawson L, Hawrysh A, Ward T, Bedard A, Sheffield BS, Lerner-Ellis J, Jacob K, Ferguson S, Kim CA, Chamberlain E, Dornan K, Waldman L, Holter S, Horte J, Hyde A, Kwon J, MacMillan A, O'Loughlin M, Tabori U, Gallinger S, Kim R. Canadian consensus for the assessment and testing of Lynch syndrome. J Med Genet 2025:jmg-2024-110465. [PMID: 40081873 DOI: 10.1136/jmg-2024-110465] [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: 10/28/2024] [Accepted: 01/11/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Lynch syndrome (LS) is an autosomal dominant cancer predisposition syndrome caused by a germline pathogenic variant, or epigenetic silencing, of a mismatch repair (MMR) gene, leading to a wide cancer spectrum with gene-specific penetrance. Ascertainment, assessment and testing of LS individuals is complex. A Canadian national guideline is needed to ensure equitable access to patient care across the country. METHODS The Canadian Lynch Syndrome (CDN-LS) working group was formed in 2021, consisting of 37 multidisciplinary LS experts and patient partners. To formulate consensus statements, a national environmental scan, Canadian clinical survey and literature review were undertaken. The e-Delphi method was used to reach consensus statements among the CDN-LS group. RESULTS The CDN-LS group voted on 21 statements, and 18 statements were adopted with over 80% agreement, including 16 statements that had over 90% agreement. These statements provide comprehensive guidelines on universal MMR reflex testing, cascade tumour testing (MLH1 promoter methylation, BRAF, somatic MMR), germline testing, therapeutics and patient advocacy. CONCLUSION This is the first comprehensive Canadian guideline for LS providing guidance to genetic specialists, laboratories, primary care providers and healthcare providers caring for patients with LS. It is endorsed by the Canadian College of Medical Genetics and the Canadian Association of Genetic Counsellors. The consensus statements are presented as a model for standard of care that improves equitable access to health services for LS across the country. Future work should include a national consensus on LS surveillance, with a goal to harmonise LS care across all provincial and territorial healthcare authorities.
Collapse
Affiliation(s)
- Melyssa Aronson
- Zane Cohen Centre for Digestive Diseases, Sinai Health System, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Laura Palma
- Specialized Medicine, Division of Medical Genetics and Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Kara Semotiuk
- Zane Cohen Centre for Digestive Diseases, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Nuk
- Hereditary Cancer Program, BC Cancer Agency, Victoria, British Columbia, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Aaron Pollett
- University of Toronto, Toronto, Ontario, Canada
- Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada
| | - Harminder Singh
- Rady Faculty of Health Sciences, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Heidi Rothenmund
- Program of Genetics and Metabolism, Shared Health Diagnostics, Winnipeg, Manitoba, Canada
- Rady Faculty of Health Sciences, Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hilary Racher
- Dynacare, Brampton, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Stephen E Pautler
- Departments of Urology and Oncology, Western University, London, Ontario, Canada
| | - Alison Rusnak
- Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Mari Rutka
- Patient Partner, Toronto, Ontario, Canada
| | - Holly Etchegary
- Clinical Epidemiology, Memorial University of Newfoundland, St John's, Newfoundland, Canada
| | | | - Pardeep Kaurah
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lesa Dawson
- Department of Obstetrics and Gynaecology, Memorial University of Newfoundland, St John's, Newfoundland, Canada
- Department of Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrea Hawrysh
- Division of Medical Genetics, Saskatchewan Health Authority, Saskatoon, Saskatchewan, Canada
| | - Thomas Ward
- Zane Cohen Centre for Digestive Diseases, Sinai Health System, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Angela Bedard
- Hereditary Cancer Program, BC Cancer Agency, Victoria, British Columbia, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Brandon S Sheffield
- Department of Laboratory Medicine, William Osler Health System, Brampton, Ontario, Canada
| | - Jordan Lerner-Ellis
- University of Toronto, Toronto, Ontario, Canada
- Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada
| | - Karine Jacob
- McGill University Health Centre, Montreal, Québec, Canada
| | - Sarah Ferguson
- Division of Gynecologic Oncology, University Health Network, Toronto, Ontario, Canada
| | - Christina A Kim
- Rady Faculty of Health Sciences, Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | | | - Kimberly Dornan
- Clinical and Metabolic Genetics Program, Hereditary Cancer Clinic, Alberta Health Services, Edmonton, Alberta, Canada
| | - Larissa Waldman
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Spring Holter
- Princess Margaret Hospital, Toronto, Ontario, Canada
| | - Janice Horte
- Medical Genetics, Edmonton Hereditary Cancer Clinic, University of Alberta Hospital, Edmonton, Alberta, Canada
- Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Angela Hyde
- Dr H Bliss Murphy Cancer Centre, St John's, Newfoundland, Canada
| | - Janice Kwon
- Department of Obstetrics and Gynaecology, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Andree MacMillan
- Provincial Medical Genetics Program, Provincial Medical Genetics Program, St John's, Newfoundland, Canada
| | - Melanie O'Loughlin
- Hereditary Cancer Program, BC Cancer Agency, Victoria, British Columbia, Canada
| | - Uri Tabori
- University of Toronto, Toronto, Ontario, Canada
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Steven Gallinger
- Wallace McCain Centre for Pancreatic Cancer, Princess Margaret Hospital, Toronto, Ontario, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Raymond Kim
- Fred A Litwin Family Centre in Genetic Medicine, University Health Network, Toronto, Ontario, Canada
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Viart NM, Renault AL, Eon-Marchais S, Jiao Y, Fuhrmann L, El Houdigui SM, Le Gal D, Cavaciuti E, Dondon MG, Beauvallet J, Raynal V, Stoppa-Lyonnet D, Vincent-Salomon A, Andrieu N, Southey MC, Lesueur F. Breast tumors from ATM pathogenic variant carriers display a specific genome-wide DNA methylation profile. Breast Cancer Res 2025; 27:36. [PMID: 40069712 PMCID: PMC11899765 DOI: 10.1186/s13058-025-01988-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The ataxia-telangiectasia mutated (ATM) kinase phosphorylates and activates several downstream targets that are essential for DNA damage repair, cell cycle inhibition and apoptosis. Germline biallelic inactivation of the ATM gene causes ataxia-telangiectasia (A-T), and heterozygous pathogenic variant (PV) carriers are at increased risk of cancer, notably breast cancer. This study aimed to investigate whether DNA methylation profiling can be useful as a biomarker to identify tumors arising in ATM PV carriers, which may help for the management and optimal tailoring of therapies of these patients. METHODS Breast tumor enriched DNA was prepared from 2 A-T patients, 27 patients carrying an ATM PV, 6 patients carrying a variant of uncertain clinical significance and 484 noncarriers enrolled in epidemiological studies conducted in France and Australia to investigate genetic and nongenetic factors involved in breast cancer susceptibility. Genome-wide DNA methylation analysis was performed using the Illumina Infinium HumanMethylation EPIC and 450K BeadChips. Correlation between promoter methylation and gene expression was assessed for 10 tumors for which transcriptomic data were available. RESULTS We found that the ATM promoter was hypermethylated in 62% of tumors of heterozygous PV carriers compared to the mean methylation level of ATM promoter in tumors of noncarriers. Gene set enrichment analyses identified 47 biological pathways enriched in hypermethylated genes involved in neoplastic, neurodegenerative and metabolic-related pathways in tumor of PV carriers. Among the 327 differentially methylated promoters, promoters of ARHGAP40, SCGB3A1 (HIN-1), and CYBRD1 (DCYTB) were hypermethylated and associated with a lower gene expression in these tumors. Moreover, using three different deep learning algorithms (logistic regression, random forest and XGBoost), we identified a set of 27 additional biomarkers predictive of ATM status, which could be used in the future to provide evidence for or against pathogenicity in ATM variant classification strategies. CONCLUSIONS We showed that breast tumors that arise in women who carry an ATM PV display a specific genome-wide DNA methylation profile. Specifically, the methylation pattern of 27 key gene promoters was predictive of ATM PV status of the women. These genes may also represent new medical prevention and therapeutic targets for these women.
Collapse
Affiliation(s)
- Nicolas M Viart
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Anne-Laure Renault
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
- Monash University, Clayton, VIC; University of Melbourne, Parkville, VIC, Australia
| | | | - Yue Jiao
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | | | | | - Dorothée Le Gal
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Eve Cavaciuti
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | | | - Juana Beauvallet
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Virginie Raynal
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL University, Paris, France
| | | | | | - Nadine Andrieu
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France
| | - Melissa C Southey
- Monash University, Clayton, VIC; University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Fabienne Lesueur
- Inserm, U1331, Institut Curie, PSL University, Mines ParisTech, Paris, France.
| |
Collapse
|
14
|
Reyneke GL, Lambert K, Beck EJ. Food-based indexes and their association with dietary inflammation. Adv Nutr 2025; 16:100400. [PMID: 40043850 DOI: 10.1016/j.advnut.2025.100400] [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: 12/04/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/23/2025] Open
Abstract
Chronic inflammation is associated with an increased risk of noncommunicable diseases, prompting an intensified interest in the diet-disease relationship for modulating inflammation. Diet quality indexes are widely used to quantify dietary patterns. However, the optimal tool for assessing dietary quality in relation to chronic inflammation remains unclear. The objective of this study was to synthesize the literature on food-based diet quality indexes and their association with chronic inflammation. A systematic scoping review of scientific databases was conducted from inception to March 2024. Studies describing the development and validation of original dietary inflammatory indexes or assessed associations between established indexes and inflammatory biomarkers were included. Studies that predominantly focused on nutrient-based indexes were excluded. Forty-three food-based indexes, evaluated across 65 studies, were categorized into 4 distinct groups based on dietary patterns (n = 18), dietary guidelines (n = 14), dietary inflammatory potential (n = 6), and therapeutic diets (n = 5). Established indexes based on the Mediterranean diet and dietary guidelines were the most extensively utilized, demonstrating inverse associations with several inflammatory biomarkers across diverse populations. The Anti-Inflammatory Diet Index, Dietary Inflammation Score, and Empirical Dietary Inflammatory Index were identified as robust, empirically derived indexes to assess diet quality based on their inflammatory potential. The dietary composition of the evaluated indexes ranged from 4 to 28 dietary components, with fruits, vegetables, whole grains, and legumes consistently classified as favorable, whereas red/processed meats and added sugars were unfavorable. This scoping review identified several promising food-based indexes for assessing inflammation-related diet quality. Methodological variations and inconsistencies in algorithms underscore the need for further validation across diverse populations. Future research should consider the scoring methods, dietary composition, and validated inflammatory biomarkers when selecting indexes to evaluate diet-inflammation associations. Understanding the characteristics that underpin these indexes informs their application in nutrition research and clinical practice.
Collapse
Affiliation(s)
- Gynette L Reyneke
- School of Health Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Kelly Lambert
- School of Medical, Indigenous, and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, Australia
| | - Eleanor J Beck
- School of Health Sciences, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, Australia; School of Medical, Indigenous, and Health Sciences, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, New South Wales, Australia.
| |
Collapse
|
15
|
Mchenga M, Vijayasingham L, RamPrakash R, Remme M. Value is Gendered: The Need for Sex and Gender Considerations in Health Economic Evaluations. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2025; 23:171-181. [PMID: 39666245 PMCID: PMC11811431 DOI: 10.1007/s40258-024-00930-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 12/13/2024]
Abstract
Economic evaluations play a crucial role in health resource allocation by assessing the costs and effects of various interventions. However, existing methodologies often overlook significant differences related to sex and gender, leading to a 'blind spot' in understanding patient heterogeneity. This paper highlights how biological and social factors influence costs and health outcomes differently for women, emphasising the need for a more explicit consideration of these differences in economic evaluations to ensure efficient and equitable resource allocation. The paper is structured to first outline how sex and gender factors impact costs and outcomes. It then identifies biases in current economic evaluation methods and practices, using real-world examples to illustrate the implications of these biases on policymaking and health equity. Notably, we argue that neglecting gender considerations can lead to inefficiencies and inequities in healthcare resource distribution. Key areas of gender bias include the estimation of productivity losses, quality of life variations and the secondary household effects of interventions. The analysis reveals that women often face higher healthcare costs and experience different health outcomes due to systemic biases in treatment and care. The paper concludes with practical recommendations for analysts, decision makers and research funders, advocating for the integration of sex and gender-responsive methodologies in health economic evaluations. Ultimately, this work calls for a paradigm shift in health economics to better reflect the complexities of sex and gender and improve health outcomes for all.
Collapse
Affiliation(s)
- Martina Mchenga
- Centre for Social Science Research, University of Cape Town, Cape Town, South Africa.
| | | | | | - Michelle Remme
- The Global Fund to Fight AIDS, Tuberculosis and Malaria, Geneva, Switzerland
- United Nations University International Institute for Global Health, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Karunaithas S, Chaibun T, Chatchawal P, Promptmas C, Buajeeb W, Yin LS, Jearanaikoon P, Lertanantawong B. Electrochemical Duplex Detection of E2 and E6 Genes of Human Papillomavirus Type 16 and Determination of Physical Status in High-Risk Cervical Carcinoma. J Med Virol 2025; 97:e70299. [PMID: 40071579 PMCID: PMC11898155 DOI: 10.1002/jmv.70299] [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: 10/28/2024] [Revised: 02/19/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Human papillomavirus type 16 (HPV-16) is a key driver in the development of cervical carcinoma, with the integration of its genome into the host DNA marking a critical step in disease progression. Monitoring the physical state of HPV-16, particularly the transition from episomal to integrated forms, is essential for evaluating the risk of malignancy development in cervix. This study presents the development of a duplex electrochemical biosensor for the simultaneous detection of the E2 and E6 genes of HPV-16. Using a one-step sandwich hybridization assay, the biosensor was able to detect HPV-16 E2 and E6 genes with a sensitivity of 8 copies/mL and 12 copies/mL respectively and distinguish between the episomal and integrated forms based on the E2/E6 ratio (cut-off 0.77, 100% sensitivity/specificity). The sensor was validated with 30 clinical cervical tissue samples, providing results comparable to qPCR method. This novel biosensor offers a rapid and efficient platform for the detection and monitoring of HPV-16, with potential applications in cervical cancer screening and prognosis.
Collapse
Affiliation(s)
- Sinthu Karunaithas
- Biosensors Laboratory, Department of Biomedical EngineeringFaculty of Engineering, Mahidol UniversityNakhon PathomThailand
| | - Thanyarat Chaibun
- Biosensors Laboratory, Department of Biomedical EngineeringFaculty of Engineering, Mahidol UniversityNakhon PathomThailand
| | - Patutong Chatchawal
- Center for Research and Development of Medical Diagnostic LaboratoriesFaculty of Associated Medical Sciences, Khon Kaen UniversityKhon KaenThailand
| | - Chamras Promptmas
- Biosensors Laboratory, Department of Biomedical EngineeringFaculty of Engineering, Mahidol UniversityNakhon PathomThailand
| | - Waranun Buajeeb
- Department of Oral Medicine and PeriodontologyFaculty of Dentistry, Mahidol UniversityBangkokThailand
| | - Lee Su Yin
- Faculty of Applied SciencesAIMST UniversityBedongMalaysia
- Centre of Excellence for Omics‐Driven Computational Biodiscovery (ComBio), AIMST UniversityBedongMalaysia
| | - Patcharee Jearanaikoon
- Center for Research and Development of Medical Diagnostic LaboratoriesFaculty of Associated Medical Sciences, Khon Kaen UniversityKhon KaenThailand
| | - Benchaporn Lertanantawong
- Biosensors Laboratory, Department of Biomedical EngineeringFaculty of Engineering, Mahidol UniversityNakhon PathomThailand
| |
Collapse
|
17
|
Sant’Angelo D, Descamps G, Lecomte V, Stanicki D, Penninckx S, Dragan T, Van Gestel D, Laurent S, Journe F. Therapeutic Approaches with Iron Oxide Nanoparticles to Induce Ferroptosis and Overcome Radioresistance in Cancers. Pharmaceuticals (Basel) 2025; 18:325. [PMID: 40143107 PMCID: PMC11945075 DOI: 10.3390/ph18030325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The emergence of nanotechnology in medicine, particularly using iron oxide nanoparticles (IONPs), may impact cancer treatment strategies. IONPs exhibit unique properties, such as superparamagnetism, biocompatibility, and ease of surface modification, making them ideal candidates for imaging, and therapeutic interventions. Their application in targeted drug delivery, especially with traditional chemotherapeutic agents like cisplatin, has shown potential in overcoming limitations such as low bioavailability and systemic toxicity of chemotherapies. Moreover, IONPs, by releasing iron ions, can induce ferroptosis, a form of iron-dependent cell death, which offers a promising pathway to reverse radio- and chemoresistance in cancer therapy. In particular, IONPs demonstrate significant potential as radiosensitisers, enhancing the effects of radiotherapy by promoting reactive oxygen species (ROS) generation, lipid peroxidation, and modulating the tumour microenvironment to stimulate antitumour immune responses. This review explores the multifunctional roles of IONPs in radiosensitisation through ferroptosis induction, highlighting their promise in advancing treatment for head and neck cancers. Additional research is crucial to fully addressing their potential in clinical settings, offering a novel approach to personalised cancer treatment.
Collapse
Affiliation(s)
- Dorianne Sant’Angelo
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Géraldine Descamps
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Valentin Lecomte
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Dimitri Stanicki
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Sébastien Penninckx
- Department of Medical Physics, Institut Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Tatiana Dragan
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Dirk Van Gestel
- Department of Radiotherapy, Institute Jules Bordet, Hopital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (T.D.); (D.V.G.)
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons (UMONS), 7000 Mons, Belgium; (G.D.); (V.L.); (D.S.); (S.L.)
| | - Fabrice Journe
- Department of Human Biology and Toxicology (Cancer Research Unit), Faculty of Medicine, Research Institute for Health Sciences and Technology, University of Mons (UMONS), 7000 Mons, Belgium
- Laboratory of Clinical and Experimental Oncology (LOCE), Institute Jules Bordet, HUB, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
18
|
Davyson E, Shen X, Huider F, Adams MJ, Borges K, McCartney DL, Barker LF, van Dongen J, Boomsma DI, Weihs A, Grabe HJ, Kühn L, Teumer A, Völzke H, Zhu T, Kaprio J, Ollikainen M, David FS, Meinert S, Stein F, Forstner AJ, Dannlowski U, Kircher T, Tapuc A, Czamara D, Binder EB, Brückl T, Kwong ASF, Yousefi P, Wong CCY, Arseneault L, Fisher HL, Mill J, Cox SR, Redmond P, Russ TC, van den Oord EJCG, Aberg KA, Penninx BWJH, Marioni RE, Wray NR, McIntosh AM. Insights from a methylome-wide association study of antidepressant exposure. Nat Commun 2025; 16:1908. [PMID: 39994233 PMCID: PMC11850842 DOI: 10.1038/s41467-024-55356-x] [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: 05/21/2024] [Accepted: 12/09/2024] [Indexed: 02/26/2025] Open
Abstract
This study tests the association of whole-blood DNA methylation and antidepressant exposure in 16,531 individuals from Generation Scotland (GS), using self-report and prescription-derived measures. We identify 8 associations and a high concordance of results between self-report and prescription-derived measures. Sex-stratified analyses observe nominally significant increased effect estimates in females for four CpGs. There is observed enrichment for genes expressed in the Amygdala and annotated to synaptic vesicle membrane ontology. Two CpGs (cg15071067; DGUOK-AS1 and cg26277237; KANK1) show correlation between DNA methylation with the time in treatment. There is a significant overlap in the top 1% of CpGs with another independent methylome-wide association study of antidepressant exposure. Finally, a methylation profile score trained on this sample shows a significant association with antidepressant exposure in a meta-analysis of eight independent external datasets. In this large investigation of antidepressant exposure and DNA methylation, we demonstrate robust associations which warrant further investigation to inform on the design of more effective and tolerated treatments for depression.
Collapse
Affiliation(s)
- E Davyson
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - X Shen
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - F Huider
- Complex Trait Genetics, Center of Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Biological Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M J Adams
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - K Borges
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - D L McCartney
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - L F Barker
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
| | - J van Dongen
- Complex Trait Genetics, Center of Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Department of Biological Psychiatry, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Research Institute, Amsterdam, The Netherlands
| | - D I Boomsma
- Complex Trait Genetics, Center of Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development, Research Institute, Amsterdam, The Netherlands
| | - A Weihs
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489, Greifswald, Germany
| | - H J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17489, Greifswald, Germany
| | - L Kühn
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475, Greifswald, Germany
| | - A Teumer
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475, Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17489, Greifswald, Germany
| | - H Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17489, Greifswald, Germany
- Department SHIP/Clinical-Epidemiological Research, Institute for Community Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - T Zhu
- Institute for Molecular Medicine Finland FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - J Kaprio
- Institute for Molecular Medicine Finland FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - M Ollikainen
- Institute for Molecular Medicine Finland FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - F S David
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
| | - S Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - F Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - A J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
- Center for Human Genetics, University of Marburg, Marburg, Germany
| | - U Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - T Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior, University of Marburg, Marburg, Germany
| | - A Tapuc
- Max Planck School of Cognition, Leipzig, Germany
- Max-Planck-Institute of Psychiatry, Department Genes and Environment, Munich, Germany
| | - D Czamara
- Max-Planck-Institute of Psychiatry, Department Genes and Environment, Munich, Germany
| | - E B Binder
- Max-Planck-Institute of Psychiatry, Department Genes and Environment, Munich, Germany
| | - T Brückl
- Max-Planck-Institute of Psychiatry, Department Genes and Environment, Munich, Germany
| | - A S F Kwong
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
| | - P Yousefi
- Medical Research Council Integrative Epidemiology Unit at the University of Bristol, University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Bristol, UK
| | - C C Y Wong
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - L Arseneault
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - H L Fisher
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- ESRC Centre for Society and Mental Health, King's College London, London, UK
| | - J Mill
- Department of Clinical & Biomedical Sciences, University of Exeter Medical School, University of Exeter, Exeter, UK
| | - S R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - P Redmond
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - T C Russ
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- Alzheimer Scotland Dementia Research Centre, University of Edinburgh, Edinburgh, UK
- Neuroprogressive and Dementia Network, NHS Research Scotland, Scotland, UK
| | - E J C G van den Oord
- Center for Biomarker Research and Precision Medicine (BPM), Virginia Commonwealth University, Virginia, USA
| | - K A Aberg
- Center for Biomarker Research and Precision Medicine (BPM), Virginia Commonwealth University, Virginia, USA
| | - B W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - R E Marioni
- Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - N R Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, 4072, Australia
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - A M McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
19
|
Marchewczyk P, Costeira B, da Silva FB, Cavadas D, Abecasis N, Limbert M, Maciel J. Quality of life outcomes in colorectal cancer survivors: insights from an observational study at a tertiary cancer center. Qual Life Res 2025:10.1007/s11136-025-03918-x. [PMID: 39966198 DOI: 10.1007/s11136-025-03918-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Colorectal cancer (CRC) significantly impacts the quality of life (QoL) of survivors, yet detailed assessments of long-term QoL are sparse. This study evaluates QoL among CRC survivors, examining the influence of different treatments and patient characteristics on outcomes. METHODS We conducted a cross-sectional study at a tertiary cancer center in Portugal, enrolling CRC patients who underwent curative surgery from 2013 to 2022. QoL was assessed using the EORTC QLQ-C30 and QLQ-CR29 at 1-, 3-, 5-, and 10-year follow-up intervals. Subgroup analyses were performed based on tumor location, radiotherapy administration, chemotherapy administration, presence of a stoma, and time since treatment, with sociodemographic and clinical factors examined on univariate and multivariate analysis. RESULTS Of the 825 eligible patients, 324 were invited and 179 participated (response rate: 55.2%). Overall, patients reported high global QoL and functional scores with low symptom scores, comparable to those of the general population. However, rectal cancer survivors experienced poorer outcomes in role and social functioning, body image, and symptom management. Those receiving radiotherapy or chemotherapy reported more symptoms, with chemotherapy recipients showing lower functional scores. Patients with a stoma had significantly lower QoL across functional and symptom scales. Long-term survivors reported decreased physical functioning. Multivariate analysis identified female gender, open surgery, and chemotherapy as factors associated with reduced QoL. CONCLUSION This study highlights significant disparities in QoL outcomes between CRC survivors, with QoL influenced by gender, cancer location, radiotherapy or chemotherapy, stoma presence, and survivorship duration, underscoring the need for personalized support programs and tailored care plans.
Collapse
Affiliation(s)
- Pola Marchewczyk
- Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
| | - Beatriz Costeira
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal
| | - Francisca Brito da Silva
- Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal
| | - Daniela Cavadas
- Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal
| | - Nuno Abecasis
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal
| | - Manuel Limbert
- Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal
| | - João Maciel
- Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisboa, 1649-028, Portugal.
- Department of General Surgery, Instituto Português de Oncologia de Lisboa Francisco Gentil, R. Prof. Lima Basto, Lisboa, 1099-023, Portugal.
| |
Collapse
|
20
|
Działach E, Malchrowicz-Mośko E, Rozmiarek M, Meller J, Juraszek P, Nowara E, Czech E, Nowaczyk P, Grajek M. COVID-19 Pandemic Conditions Affecting QoL and Mental Health of Oncology Patients in Poland. Cancers (Basel) 2025; 17:662. [PMID: 40002257 PMCID: PMC11852427 DOI: 10.3390/cancers17040662] [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: 01/01/2025] [Revised: 02/07/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The COVID-19 pandemic has caused widespread disruptions in oncology care, significantly affecting both the quality of life (QoL) and mental health of cancer patients. This study aimed to evaluate the long-term impacts of the pandemic on oncology patients, focusing on the periods before, during, and after the pandemic. Objective: The objective of this study was to assess the changes in QoL, illness acceptance, and mental health indicators, including the risk of depression, generalized anxiety, elevated stress levels, and post-traumatic stress disorder (PTSD) among oncology patients, comparing these factors across the pre-pandemic, pandemic, and post-pandemic periods. Material and Methods: This study included 2000 oncology patients, divided into three cohorts based on the time of assessment: pre-pandemic (2019, n = 600), during the pandemic (2020-2021, n = 800), and post-pandemic (2023, n = 600). This study included a balanced sample of 52% female and 48% male participants, with a mean age of 58 years (SD = 11.9), representing a wide range of cancer types including breast (25.7%), lung (20.9%), and colorectal cancer (14.8%). Additional demographics showed a mean BMI of 25.8, with varied educational levels, marital statuses, income levels, and lifestyle factors such as smoking and alcohol consumption. QoL was assessed using the EORTC QLQ-C30, while the Hospital Anxiety and Depression Scale (HADS) was used to measure depression and anxiety. The PTSD Checklist for DSM-5 (PCL-5) was used to evaluate PTSD symptoms, and stress levels were measured with the Perceived Stress Scale (PSS). Statistical analyses were conducted using ANOVA and chi-square tests to assess differences between the groups. Results: During the pandemic, the prevalence of depression symptoms rose significantly, from 15% pre-pandemic to 32% (p < 0.001), while the risk of generalized anxiety increased from 18% to 40% (p < 0.001). Stress levels also saw a sharp rise, with 45% of patients reporting elevated stress during the pandemic compared to 22% before (p < 0.001). The rate of PTSD symptoms increased from 10% pre-pandemic to 28% during the pandemic (p < 0.001). QoL scores dropped markedly, with the mean EORTC QLQ-C30 global health status score decreasing by 25% during the pandemic (p < 0.01). Illness acceptance declined, with 60% of patients reporting poor acceptance during the pandemic, compared to 35% before. In the post-pandemic period, a slight improvement was observed across all measures. Depression levels dropped to 28% (p < 0.05 compared to the pandemic period), and anxiety levels decreased to 35% (p < 0.05). Stress and PTSD symptoms also showed modest reductions, with 38% reporting elevated stress and 22% exhibiting PTSD symptoms (p < 0.05). However, these post-pandemic values remained significantly higher than pre-pandemic levels (p < 0.001). QoL improved marginally, with a 10% increase in the global health status score compared to the pandemic period, though it remained lower than pre-pandemic scores (p < 0.05). Conclusions: The COVID-19 pandemic had a profound impact on the mental health and QoL of oncology patients, with significant increases in depression, anxiety, stress, and PTSD symptoms, along with a decrease in QoL and illness acceptance. While post-pandemic recovery trends are apparent, the psychological burden remains elevated compared to pre-pandemic conditions. These findings highlight the need for continued mental health support and interventions for oncology patients, even after the immediate pandemic effects have subsided.
Collapse
Affiliation(s)
- Eliza Działach
- Department of Public Health, Faculty of Public Health in Bytom, Silesian Medical University in Katowice, 41-902 Bytom, Poland; (E.D.); (P.J.); (M.G.)
| | - Ewa Malchrowicz-Mośko
- Department of Tourism and Recreation, Faculty of Physical Education, Józef Piłsudski University of Physical Education, 00-968 Warsaw, Poland
| | - Mateusz Rozmiarek
- Department of Sports Tourism, Faculty of Physical Culture Sciences, Poznan University of Physical Education, 61-871 Poznan, Poland;
| | - Jolanta Meller
- Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland;
| | - Paweł Juraszek
- Department of Public Health, Faculty of Public Health in Bytom, Silesian Medical University in Katowice, 41-902 Bytom, Poland; (E.D.); (P.J.); (M.G.)
| | - Elżbieta Nowara
- Department of Health Sciences, Jan Dlugosz University of Humanities and Natural Sciences, 42-200 Czestochowa, Poland;
| | - Elżbieta Czech
- Department of Epidemiology and Biostatistics, Faculty of Public Health in Bytom, Silesian Medical University in Katowice, 41-902 Bytom, Poland;
| | - Piotr Nowaczyk
- Breast Surgical Oncology Department, Breast Cancer Unit, Greater Poland Cancer Center, Garbary 15, 61-866 Poznan, Poland;
| | - Mateusz Grajek
- Department of Public Health, Faculty of Public Health in Bytom, Silesian Medical University in Katowice, 41-902 Bytom, Poland; (E.D.); (P.J.); (M.G.)
| |
Collapse
|
21
|
Chan JA, Geyer S, Zemla T, Knopp MV, Behr S, Pulsipher S, Ou FS, Dueck AC, Acoba J, Shergill A, Wolin EM, Halfdanarson TR, Konda B, Trikalinos NA, Tawfik B, Raj N, Shaheen S, Vijayvergia N, Dasari A, Strosberg JR, Kohn EC, Kulke MH, O'Reilly EM, Meyerhardt JA. Phase 3 Trial of Cabozantinib to Treat Advanced Neuroendocrine Tumors. N Engl J Med 2025; 392:653-665. [PMID: 39282913 PMCID: PMC11821447 DOI: 10.1056/nejmoa2403991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
BACKGROUND Treatment options for patients with advanced neuroendocrine tumors are limited. The efficacy of cabozantinib in the treatment of previously treated, progressive extrapancreatic or pancreatic neuroendocrine tumors is unclear. METHODS We enrolled two independent cohorts of patients - those with extrapancreatic neuroendocrine tumors and those with pancreatic neuroendocrine tumors - who had received peptide receptor radionuclide therapy or targeted therapy or both. Patients were randomly assigned in a 2:1 ratio to receive cabozantinib at a dose of 60 mg daily or placebo. The primary end point was progression-free survival as assessed by blinded independent central review. Key secondary end points included objective response, overall survival, and safety. RESULTS In the cohort of 203 patients with extrapancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 8.4 months, as compared with 3.9 months with placebo (stratified hazard ratio for progression or death, 0.38; 95% confidence interval [CI], 0.25 to 0.59; P<0.001). In the cohort of 95 patients with pancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 13.8 months, as compared with 4.4 months with placebo (stratified hazard ratio, 0.23; 95% CI, 0.12 to 0.42; P<0.001). The incidence of confirmed objective response with cabozantinib was 5% and 19% among patients with extrapancreatic and pancreatic neuroendocrine tumors, respectively, as compared with 0% with placebo. Grade 3 or higher adverse events were noted in 62 to 65% of the patients treated with cabozantinib, as compared with 23 to 27% of the patients who received placebo. Common treatment-related adverse events of grade 3 or higher included hypertension, fatigue, diarrhea, and thromboembolic events. CONCLUSIONS Cabozantinib, as compared with placebo, significantly improved progression-free survival in patients with previously treated, progressive advanced extrapancreatic or pancreatic neuroendocrine tumors. Adverse events were consistent with the known safety profile of cabozantinib. (Funded by the National Cancer Institute and others; CABINET ClinicalTrials.gov number, NCT03375320.).
Collapse
Affiliation(s)
- Jennifer A Chan
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Susan Geyer
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Tyler Zemla
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Michael V Knopp
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Spencer Behr
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Sydney Pulsipher
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Fang-Shu Ou
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Amylou C Dueck
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Jared Acoba
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Ardaman Shergill
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Edward M Wolin
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Thorvardur R Halfdanarson
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Bhavana Konda
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Nikolaos A Trikalinos
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Bernard Tawfik
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Nitya Raj
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Shagufta Shaheen
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Namrata Vijayvergia
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Arvind Dasari
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Jonathan R Strosberg
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Elise C Kohn
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Matthew H Kulke
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Eileen M O'Reilly
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| | - Jeffrey A Meyerhardt
- From Dana-Farber Cancer Institute (J.A.C., J.A.M.), Boston Medical Center (M.H.K.), and Boston University (M.H.K.) - all in Boston; the Alliance Statistics and Data Management Center, Mayo Clinic (S.G., T.Z., S.P., F.-S.O.), and Mayo Clinic Comprehensive Cancer Center (T.R.H.) - both in Rochester, MN; Wright Center of Innovation and the Imaging and Radiation Oncology Core, University of Cincinnati, Cincinnati (M.V.K.), and the Ohio State University Comprehensive Cancer Center, Columbus (B.K.) - both in Ohio; the University of California, San Francisco, San Francisco (S.B.), and Stanford Cancer Center, Stanford (S.S.) - both in California; Alliance Statistics and Data Management Center, Mayo Clinic, Scottsdale, AZ (A.C.D.); the University of Hawaii Cancer Center, Honolulu (J.A.); the Alliance Protocol Operations Office, University of Chicago, Chicago (A.S.); Mount Sinai Medical Center (E.M.W.) and Memorial Sloan Kettering Cancer Center (N.R., E.M.O.) - both in New York; Washington University School of Medicine and Siteman Cancer Center, St. Louis (N.A.T.); the University of New Mexico Comprehensive Cancer Center, Albuquerque (B.T.); Fox Chase Cancer Center, Philadelphia (N.V.); M.D. Anderson Cancer Center, Houston (A.D.); Moffitt Cancer Center, Tampa, FL (J.R.S.); and the National Cancer Institute, Bethesda, MD (E.C.K.)
| |
Collapse
|
22
|
Fountoukidis G, Schiza A, Smith D, Othman M, Bergman M, Ahlgren J, Lambe M, Irenaeus S, Valachis A. Effect of alcohol consumption on oncological treatment effectiveness and toxicity in patients with cancer: a systematic review and meta-analysis. BMC Cancer 2025; 25:246. [PMID: 39939877 PMCID: PMC11823036 DOI: 10.1186/s12885-025-13694-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 02/10/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Alcohol consumption has been associated with an increased risk of cancer-related mortality. It may also negatively impact oncological therapies, potentially leading to impaired effectiveness or an increased risk of treatment-related toxicities. The aim of this systematic review and meta-analysis was to examine the current evidence regarding the potential effects of alcohol consumption during cancer treatments on both treatment effectiveness and toxicity, irrespective of cancer type. METHODS A comprehensive literature search was performed across three electronic databases (Medline, Web of Science, Cochrane) covering studies from January 1990 to December 2023. Furthermore, a manual search based on the reference lists of the eligible studies was performed to identify additional potentially eligible studies. Studies were eligible if they involved cancer patients and provided data on alcohol consumption during specific oncological treatments, including its effect on treatment outcomes, or compared treatment effectiveness or toxicity between drinkers and non-drinkers. Studies were excluded if they did not meet these criteria, were duplicates, case reports, conference abstracts, or focused only on cancer-specific or overall survival. Only studies using multivariable analyses to examine the association between alcohol consumption and treatment effectiveness or toxicity were included in the pooled analyses. Pooled Hazard Ratios (HRs) or Odds Ratios (ORs) and their corresponding 95% Confidence Intervals (CIs) were calculated using random-effects models. Study quality was assessed by using the Newcastle-Ottawa scale whereas the GRADE approach was applied to rate the certainty of evidence for pooled analyses. RESULTS Out of 6734 studies identified through searching, 38 met the inclusion criteria for pooled analyses. Alcohol consumption during radiotherapy, with or without concomitant chemotherapy, was associated with worse disease-free survival (pooled HR: 2.05; 95% CI: 1.09 - 3.89), although the numerically increased risk for locoregional recurrence did not reach statistically significance (pooled HR: 2.01; 95% CI: 0.76 - 5.36). The potential impact of alcohol consumption on chemotherapy-induced neurotoxicity and acute / delayed nausea was not statistically significant. However, alcohol consumption was associated with a lower risk of overall chemotherapy-induced nausea (OR: 0.69; 95% CI: 0.57, 0.84). CONCLUSION Our findings suggest that alcohol consumption may have a negative impact on radiotherapy, whereas its potential impact on the effectiveness of systemic oncological therapies (chemotherapy, molecular targeted therapy, immunotherapy, endocrine therapy) has not been adequately studied. Similarly, the current evidence on the potential association between alcohol consumption and treatment-related toxicities is weak, highlighting the need for well-designed prospective studies on this topic.
Collapse
Affiliation(s)
- Georgios Fountoukidis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, 702 81, Sweden
| | - Aglaia Schiza
- Department of Oncology, Uppsala University Hospital, Uppsala, 751 85, Sweden.
- Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Uppsala, Sweden.
| | - Daniel Smith
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Örebro University, Örebro, 702 81, Sweden
| | - Mukhrizah Othman
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, 702 81, Sweden
| | - Marie Bergman
- Department of Oncology, Hospital of Karlstad, Karlstad, 652 30, Sweden
| | - Johan Ahlgren
- Regional Cancer Centre, Mid-Sweden Health Care Region, Uppsala, 751 22, Sweden
| | - Mats Lambe
- Regional Cancer Centre, Mid-Sweden Health Care Region, Uppsala, 751 22, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, 104 35, Sweden
| | - Sandra Irenaeus
- Regional Cancer Centre, Mid-Sweden Health Care Region, Uppsala, 751 22, Sweden
| | - Antonis Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, 702 81, Sweden
| |
Collapse
|
23
|
Shawky M, Choudhary C, Coleridge SL, Bryant A, Morrison J. Neoadjuvant chemotherapy before surgery versus surgery followed by chemotherapy for initial treatment in advanced epithelial ovarian cancer. Cochrane Database Syst Rev 2025; 2:CD005343. [PMID: 39927569 PMCID: PMC11808835 DOI: 10.1002/14651858.cd005343.pub7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
RATIONALE Epithelial ovarian cancer (EOC) presents at an advanced stage in the majority of women. These women require a combination of surgery and chemotherapy for optimal treatment. Conventional treatment has been to perform surgery first and then give chemotherapy. However, there may be advantages to using chemotherapy before surgery. OBJECTIVES To assess the advantages and disadvantages of treating women with advanced EOC with chemotherapy before cytoreductive surgery (neoadjuvant chemotherapy (NACT)) compared with conventional treatment where chemotherapy follows cytoreductive surgery (primary cytoreductive surgery (PCRS)). SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, Embase, ClinicalTrials.gov, and the World Health Organization International Clinical Trials Registry Platform on 21 March 2024. We also checked the reference lists of relevant papers for further studies. We contacted the principal investigators of relevant trials for further information. ELIGIBILITY CRITERIA Randomised controlled trials (RCTs) of women with advanced epithelial ovarian cancer (International Federation of Gynecology and Obstetrics (FIGO) stage III/IV) who were randomly allocated to treatment groups that compared platinum-based chemotherapy before cytoreductive surgery with platinum-based chemotherapy following cytoreductive surgery. OUTCOMES We extracted data on overall (OS) and progression-free survival (PFS), adverse events, surgically related mortality and morbidity, and quality of life outcomes. RISK OF BIAS We used the Cochrane RoB 1 tool to assess risk of bias in RCTs. SYNTHESIS METHODS We conducted meta-analyses using random-effects models (due to heterogeneity between studies) to calculate hazard ratios (HR), risk ratios (RR), mean differences (MD), and 95% confidence intervals (CI) for all outcomes. We assessed the certainty of evidence according to the GRADE approach. INCLUDED STUDIES We identified a further 1022 titles and abstracts through our searches in this update (958 unique records after further de-duplication), adding to the 2227 titles and abstracts identified in previous versions of this review. A total of five RCTs of varying quality and size met the inclusion criteria. We identified no new completed studies in this update, but we did include additional data from existing studies. The studies assessed a total of 1774 women with stage III/IV ovarian cancer randomised to NACT followed by interval cytoreductive surgery (ICRS) or PCRS followed by chemotherapy. We included data from four studies in the meta-analyses (1692 participants). SYNTHESIS OF RESULTS Survival We found little or no difference between groups in OS (HR 0.96, 95% CI 0.86 to 1.08; P = 0.49; I2 = 0%; 4 studies; 1692 women; high-certainty evidence) and likely little or no difference between groups in PFS (HR 0.98, 95% CI 0.88 to 1.08; P = 0.62; I2 = 0%; 4 studies; 1692 women; moderate-certainty evidence). Adverse events Adverse events, surgical morbidity, and quality of life outcomes were variably and incompletely reported across studies. NACT reduces postoperative mortality (0.4% in the NACT group versus 3.3% in the PCRS group) (RR 0.18, 95% CI 0.06 to 0.52; P = 0.002; I2 = 0%; 4 studies; 1542 women; high-certainty evidence). There are probably clinically meaningful differences in favour of NACT compared to PCRS in overall surgically related adverse effects (grade 3+ (G3+)) (6% in the NACT group versus 29% in the PCRS group) (RR 0.22, 95% CI 0.13 to 0.38; P < 0.001; I2 = 0%; 2 studies; 435 women; moderate-certainty evidence). Organ resection NACT probably results in a large reduction in the need for stoma formation (5.8% in the NACT group versus 20.4% in the PCRS group) (RR 0.29, 95% CI 0.12 to 0.74; P = 0.009; I2 = 70%; 2 studies; 632 women; moderate-certainty evidence) and probably reduces the risk of needing bowel resection at the time of surgery (13.0% in the NACT group versus 26.6% in the PCRS group) (RR 0.47, 95% CI 0.27 to 0.81; P = 0.007; I2 = 84%; 4 studies; 1578 women; moderate-certainty evidence). Quality of life Global quality of life on the EORTC QLQ-C30 produced imprecise results in three studies, with high levels of heterogeneity (quality of life at six months: MD 6.62, 95% CI -2.89 to 16.13; P = 0.17; I2 = 92%; 3 studies; 559 women; low-certainty evidence). Overall, functional and symptom scores may be slightly improved for NACT at 6 months, but similar by 12 months, although the differences might not be clinically meaningful. AUTHORS' CONCLUSIONS The available high- to moderate-certainty evidence shows there is likely little or no difference in primary survival outcomes between PCRS and NACT for those with advanced EOC who are suitable for either treatment option. NACT reduces the risk of postoperative mortality and likely reduces the risk of serious adverse events, especially those around the time of surgery, and the need for stoma formation. These data should inform women and clinicians (involving specialist gynaecological multidisciplinary teams) and allow treatment to be tailored to the individual patient, taking into account surgical resectability, age, histology, stage, and performance status. Data from an unpublished study and ongoing studies are awaited. FUNDING This Cochrane review update had no dedicated funding. REGISTRATION Protocol (2005): DOI: 10.1002/14651858.CD005343 Original review (2007): DOI: 10.1002/14651858.CD005343.pub2 Review update (2012): DOI: 10.1002/14651858.CD005343.pub3 Review update (2019): DOI: 10.1002/14651858.CD005343.pub4 Review update (2021): DOI: 10.1002/14651858.CD005343.pub5 Review updated (2021a): DOI: 10.1002/14651858.CD005343.pub6.
Collapse
Affiliation(s)
- Mohamed Shawky
- Department of Gynaecological Oncology, GRACE Centre, Musgrove Park Hospital, Somerset NHS Foundation Trust, Taunton, TA1 5DA, Somerset, UK
| | - Cherry Choudhary
- Medicine for the Elderly Department, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sarah L Coleridge
- Department of Obstetrics and Gynaecology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Andrew Bryant
- Institute of Health & Society, Newcastle University, Newcastle upon Tyne, UK
| | - Jo Morrison
- Department of Gynaecological Oncology, GRACE Centre, Musgrove Park Hospital, Somerset NHS Foundation Trust, Taunton, TA1 5DA, Somerset, UK
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
24
|
Rabbani SA, Patni MA, El-Tanani M, Rangraze IR, Wali AF, Babiker R, Satyam SM, El-Tanani Y, Almetwally AAMS. Impact of Lifestyle Modifications on Cancer Mortality: A Systematic Review and Meta-Analysis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:307. [PMID: 40005424 PMCID: PMC11857246 DOI: 10.3390/medicina61020307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Cancer survival poses significant challenges in oncology, with lifestyle modifications increasingly recognized as crucial in modifying patient outcomes post-diagnosis. This meta-analysis aims to systematically evaluate the impact of various lifestyle interventions on cancer survival across different types of cancer. Methods: A comprehensive literature search of electronic databases including PubMed, Scopus and Cochrane was performed to identify relevant studies up to 30 November 2024. Relevant studies were chosen and data were extracted and analyzed using SPSS Version 29.0 software. Results: Our systematic review included data from 98 studies involving a total of 1,461,834 cancer patients to evaluate the impact of lifestyle factors on cancer survival. Out of these, 64 studies were included in the meta-analysis. Our meta-analysis demonstrates that adherence to specific dietary patterns significantly improves cancer-specific outcomes. The Healthy Eating Index (HEI) diet was associated with a reduction in cancer-specific mortality (pooled log HR: -0.22; 95% CI: [-0.32, -0.12]; p < 0.001). Similar benefits were observed with the Mediterranean diet (aMED), which also reduced cancer mortality and recurrence (pooled log HR: -0.24; 95% CI: [-0.40, -0.07]; p < 0.001), and the Dietary Approaches to Stop Hypertension (DASH) diet (pooled log HR: -0.22; 95% CI: [-0.33, -0.12]; p < 0.001). Additionally, general dietary improvements were beneficial for breast cancer-specific mortality across 17 cohort studies (pooled log HR: -0.15; 95% CI: [-0.25, -0.06]; p < 0.001). Engaging in any form of physical activity post-diagnosis was associated with significant improvements in cancer-specific mortality or recurrence (pooled log HR: -0.31; 95% CI: [-0.38, -0.25]; p < 0.001). Participants who ceased smoking after diagnosis exhibited more favorable cancer outcomes (pooled log HR: -0.33; 95% CI: [-0.42, -0.24]; p < 0.001), with smoking cessation notably reducing cancer-specific mortality among lung cancer survivors (pooled log HR: -0.34; 95% CI: [-0.48, -0.20]; p < 0.001). Additionally, reducing alcohol intake post-diagnosis significantly improved cancer outcomes (pooled log HR: -0.26; 95% CI: [-0.33, -0.19]; p < 0.001). Alcohol moderation in gastrointestinal tract cancer survivors specifically decreased both cancer-specific mortality and recurrence (pooled log HR: -0.22; 95% CI: [-0.29, -0.15]; p < 0.001). Conclusions: Lifestyle modifications after cancer diagnosis significantly improve cancer-specific outcomes. Specific dietary patterns, increased physical activity, smoking cessation, and reduced alcohol intake are all associated with lower cancer-specific mortality. Integrating these lifestyle changes into oncology care may enhance patient survival and quality of life.
Collapse
Affiliation(s)
- Syed Arman Rabbani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Mohamed Anas Patni
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Imran Rashid Rangraze
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Adil Farooq Wali
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Rasha Babiker
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | - Shakta Mani Satyam
- Translational and Medical Research Centre, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
- RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah 11172, United Arab Emirates
| | | | | |
Collapse
|
25
|
Holthuijsen DDB, Rijnhart JJM, Bours MJL, van Roekel EH, Ueland PM, Breukink SO, Janssen-Heijnen MLG, Konsten JL, Keulen ETP, McCann A, Brezina S, Gigic B, Ulrich CM, Weijenberg MP, Eussen SJPM. Longitudinal associations of dietary intake with fatigue in colorectal cancer survivors up to 1 year post-treatment, and the potential mediating role of the kynurenine pathway. Brain Behav Immun 2025; 126:144-159. [PMID: 39922470 DOI: 10.1016/j.bbi.2025.02.003] [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: 11/14/2024] [Revised: 01/22/2025] [Accepted: 02/01/2025] [Indexed: 02/10/2025] Open
Abstract
INTRODUCTION A healthy diet may help to reduce cancer-related fatigue, but evidence is limited and mechanisms remain unclear. Both diet and fatigue following colorectal cancer (CRC) have been linked to metabolites (kynurenines) of the kynurenine pathway (KP). We investigated longitudinal associations between dietary intake and fatigue, and the potential mediating role of the KP, in CRC survivors up to 1 year post-treatment. METHODS Measurements at 6 weeks, 6 months, and 1 year post-treatment were performed in 209 stage I-III CRC survivors. Diet was assessed by 7-day food records. Plasma kynurenines were analyzed using LC-MS/MS. Fatigue, including subjective fatigue, was assessed using validated questionnaires. To analyse longitudinal associations between diet and fatigue and to explore potential mediation by the KP, we used confounder-adjusted multilevel parallel-multiple mediator models with all kynurenines included simultaneously, and simple mediator models with established KP ratios to estimate total (c: diet-fatigue), direct (c': diet-fatigue, while controlling for mediators), metabolite-specific indirect (ab: diet-metabolite-fatigue), and total indirect (ab: diet-metabolites-fatigue) effects. RESULTS Higher intake of total carbohydrates and mono- and disaccharides was longitudinally associated with more subjective fatigue, while higher intake of plant protein, total fat, and unsaturated fats was associated with less subjective fatigue (c). Most associations remained statistically significant after controlling for KP metabolites, except for mono- and disaccharides (c'). All kynurenines simultaneously did not mediate longitudinal associations between diet and subjective fatigue (ab). The kynurenic acid-to-quinolinic acid (KA/QA) ratio significantly mediated associations of intakes of carbohydrate, mono- and disaccharides, alcohol, magnesium, and zinc with subjective fatigue, whereas the HKr significantly mediated the association between polysaccharide intake and subjective fatigue (ab). CONCLUSION Our findings suggest that carbohydrate intake is associated with greater fatigue, while protein and fat intake are associated with lower fatigue in CRC survivors up to 1 year post-treatment. While all KP metabolites simultaneously did not significantly mediate associations between diet and fatigue in our population, the KA/QA ratio and HKr were significant mediators in several diet-fatigue associations. These results should be repeated in larger observational studies.
Collapse
Affiliation(s)
- Daniëlle D B Holthuijsen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | | | - Martijn J L Bours
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Eline H van Roekel
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | | | - Stéphanie O Breukink
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Surgery, Maastricht University Medical Centre+, Maastricht, the Netherlands; NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre+, Maastricht, the Netherlands
| | - Maryska L G Janssen-Heijnen
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands; Department of Clinical Epidemiology, VieCuri Medical Centre, Venlo, the Netherlands
| | - Joop L Konsten
- Department of Surgery, VieCuri Medical Centre, Venlo, the Netherlands
| | - Eric T P Keulen
- Department of Internal Medicine and Gastroenterology, Zuyderland Medical Centre Sittard-Geleen, Geleen, the Netherlands
| | | | - Stefanie Brezina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Biljana Gigic
- Department of General Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Cornelia M Ulrich
- Huntsman Cancer Institute and Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Matty P Weijenberg
- Department of Epidemiology, GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Simone J P M Eussen
- Department of Epidemiology, CARIM Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Epidemiology, CAPHRI Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
26
|
Jung H, Choi Y, Kim B. A comparative study of health behaviors in adult male cancer survivors and the general male population in Korea: from the Korea national health and nutrition examination survey VII-VIII (2016-2021). Support Care Cancer 2025; 33:160. [PMID: 39915300 PMCID: PMC11802672 DOI: 10.1007/s00520-025-09200-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND As healthy living becomes crucial for cancer survivors, discussing the health risk behaviors of male cancer survivors, who are more prone to such behaviors, is essential. This study compared health-related behaviors and obesity among male cancer survivors and the general male population in Korea. METHODS This cross-sectional, matched case-control study used data from the Korea National Health and Nutrition Examination Survey (KNHANES). Of 11,760 participants, 349 cancer survivors and 1,047 controls were matched by propensity scores. Logistic regression evaluated differences in BMI, smoking, alcohol consumption, physical activity, and diet. RESULTS Cancer survivors had lower odds of being overweight and higher odds of being former smokers and drinkers than controls. No significant differences were found in physical activity or diet. However, the middle-aged and older cancer survivors were more likely to be overweight and obese, respectively. The middle-aged survivors were also more likely to be former smokers, while the older survivors were more likely to be former drinkers. CONCLUSION Cancer survivors were more likely to have a normal weight, be past smokers, or be former drinkers compared with controls. Education on cancer prevention is required to improve health-related behaviors and prevent secondary cancer.
Collapse
Affiliation(s)
- Hyein Jung
- Division of Cancer Prevention, National Cancer Control Institute, National Cancer Center, Goyang, Gyeonggi-Do, Republic of Korea
| | - Yoonjoo Choi
- Division of Cancer Prevention, National Cancer Control Institute, National Cancer Center, Goyang, Gyeonggi-Do, Republic of Korea
| | - Byungmi Kim
- Division of Cancer Prevention, National Cancer Control Institute, National Cancer Center, Goyang, Gyeonggi-Do, Republic of Korea.
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Gyeonggi-Do, Republic of Korea.
| |
Collapse
|
27
|
Law L, Heerey JJ, Devlin BL, Brukner P, De Livera AM, Attanayake A, Cooper I, Donato A, Hebert JR, Price S, White NP, Culvenor AG. The eFEct of an Anti-Inflammatory Diet for Knee oSTeoarthritis (FEAST) Trial: Baseline Characteristics and Relationships With Dietary Inflammatory Index. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025:1-10. [PMID: 39912561 DOI: 10.1080/27697061.2025.2461219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/07/2025]
Abstract
OBJECTIVE This study aims to: (i) describe the results of recruitment into the eFfEct of an Anti-inflammatory diet for knee oSTeoarthritis (FEAST) randomized controlled trial (RCT); (ii) report baseline characteristics of randomized participants and compare these with four large international cohorts; and (iii) explore cross-sectional associations between dietary inflammatory index (DII®) scores and baseline clinical characteristics. METHODS The FEAST RCT compares an anti-inflammatory dietary programme and standard care low-fat dietary programme for adults aged 45-85 years with knee osteoarthritis (OA). At baseline, participants provided medical history (medical comorbidities, symptomatic musculoskeletal sites), completed questionnaires (demographic characteristics, Knee injury and OA Outcome Score (KOOS)) and a 3-day food diary. Both DII® and energy-adjusted DII (E-DIITM) scores were calculated based on 3-day food diary data and was used to quantify the effect of diet on systemic inflammation. Associations between DII/E-DII and KOOS subscales, symptomatic musculoskeletal sites, and comorbidities was assessed using linear and negative binomial regression. RESULTS 1121 individuals were screened to identify 182 eligible individuals, from which 144 participants (64% female, 36% male) enrolled, with a mean ± SD age 65 ± 8 years and body mass index 30.3 ± 6.2 kg/m2. Overweight (41%) and obesity (45%) was common. Two-thirds (62%) had ≥1 medical comorbidity, most commonly hypertension (26%). Musculoskeletal pain in sites other than the index knee was reported in 79%, most commonly in the lower back (42%). Mean DII and E-DII scores were 0.58 ± 1.49 and -0.31 ± 1.41, respectively. No associations were found between DII/E-DII and KOOS subscales except for activities of daily living (ADL), number of medical comorbidities and symptomatic MSK sites, and BMI. CONCLUSION The FEAST cohort is comparable to other knee OA cohorts, supporting generalizability of the results. Despite a relatively pro-inflammatory diet at baseline, DII/E-DII was not associated with KOOS subscales, number of comorbidities or symptomatic musculoskeletal sites.
Collapse
Affiliation(s)
- Lynette Law
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - Joshua J Heerey
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - Brooke L Devlin
- School of Human Movement and Nutrition Sciences, University of Queensland, Brisbane, Australia
| | - Peter Brukner
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - Alysha M De Livera
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
- Mathematics and Statistics, School of Computing, Engineering and Mathematical Sciences, La Trobe University, Bundoora, Australia
| | - Amanda Attanayake
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - Indiana Cooper
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - Amy Donato
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| | - James R Hebert
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | - Sherry Price
- Cancer Prevention and Control Program, Department of Epidemiology and Biostatistics, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina, USA
| | | | - Adam G Culvenor
- La Trobe Sport and Exercise Medicine Research Centre, School of Allied Health, Human Services and Sport, La Trobe University, Bundoora, Australia
| |
Collapse
|
28
|
Nagle CM, Ibiebele TI, Na R, Bandera EV, Cramer D, Doherty JA, Giles GG, Goodman MT, Hanley GE, Harris HR, Jensen A, Kjaer SK, Lee A, McGuire V, Milne RL, Qin B, Richardson J, Sasamoto N, Schildkraut JM, Sieh W, Terry KL, Titus L, Trabert B, Wentzensen N, Wu AH, Berchuck A, Pike MC, Pearce CL, Webb PM. Diet and survival after a diagnosis of ovarian cancer: a pooled analysis from the Ovarian Cancer Association Consortium. Am J Clin Nutr 2025:S0002-9165(25)00072-3. [PMID: 39921094 DOI: 10.1016/j.ajcnut.2025.02.004] [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: 10/13/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Prognosis after a diagnosis of invasive epithelial ovarian cancer is poor. Some studies have suggested modifiable behaviors, like diet, are associated with survival but the evidence is inconsistent. OBJECTIVES This study aims to pool data from studies conducted around the world to evaluate the relationships among dietary indices, foods, and nutrients from food sources and survival after a diagnosis of ovarian cancer. METHODS This analysis from the Multidisciplinary Ovarian Cancer Outcomes Group within the Ovarian Cancer Association Consortium included 13 studies with 7700 individuals with ovarian cancer, who completed food-frequency questionnaires regarding their prediagnosis diet. Adjusted hazard ratios (aHRs) and 95% confidence intervals (CI) for associations with overall survival were estimated using Cox proportional hazards models. RESULTS Overall, there was no association between any of the 7 dietary indices (representing prediagnosis diet) evaluated and survival; however, associations differed by tumor stage. Although there were no consistent associations among those with advanced disease, among those with earlier stage (local/regional) disease, higher scores on the alternate Healthy Eating Index (aHR quartile 4 compared with 1 = 0.66, 95% CI: 0.50, 0.87), Healthy Eating Index-2015 (aHR: 0.75; 95% CI: 0.59, 0.97), and alternate Mediterranean diet (aHR: 0.76; 95% CI: 0.60, 0.98) were associated with better survival. Better survival was also observed for individuals with early-stage disease who reported higher intakes of dietary components that contribute to the healthy diet indices (aHR for Q4 compared with Q1: vegetables 0.71; 95% CI: 0.56, 0.91), tomatoes (aHR: 0.72; 95% CI: 0.57, 0.91) and nuts and seeds (aHR 0.71; 95% CI: 0.55, 0.92). In contrast, there were suggestions of worse survival with higher scores on 2 of the 3 inflammatory indices and higher intake of trans-fatty acids. CONCLUSIONS Adherence to a more healthy, less-inflammatory diet may confer a survival benefit for individuals with early-stage ovarian cancer.
Collapse
Affiliation(s)
- Christina M Nagle
- Gynaecological Cancers Group, Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Torukiri I Ibiebele
- Gynaecological Cancers Group, Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Renhua Na
- Gynaecological Cancers Group, Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Elisa V Bandera
- Cancer Prevention and Control Program, Rutgers Cancer Institute, New Brunswick, NJ, United States
| | - Daniel Cramer
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Jennifer A Doherty
- Huntsman Cancer Institute, Department of Population Health Sciences, University of Utah, Salt Lake City, UT, United States
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Marc T Goodman
- Cancer Prevention and Control Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Community and Population Health Research Institute, Los Angeles, CA, United States
| | - Gillian E Hanley
- Department of Obstetrics & Gynaecology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Holly R Harris
- Program in Epidemiology, Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, United States; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, United States
| | - Allan Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark
| | - Susanne K Kjaer
- Department of Virus, Lifestyle and Genes, Danish Cancer Institute, Copenhagen, Denmark; Department of Gynecology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Alice Lee
- Department of Public Health, California State University, Fullerton, Fullerton, CA, United States
| | - Valerie McGuire
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, United States
| | - Roger L Milne
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, Australia
| | - Bo Qin
- Cancer Prevention and Control Program, Rutgers Cancer Institute, New Brunswick, NJ, United States
| | - Jean Richardson
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States; Patient advocate
| | - Naoko Sasamoto
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Joellen M Schildkraut
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Weiva Sieh
- Department of Epidemiology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kathryn L Terry
- Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics and Gynecology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Linda Titus
- Dartmouth Cancer Center, Lebanon, NH03756, United States
| | - Britton Trabert
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States; Department of Obstetrics and Gynecology, University of Utah, Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Nicolas Wentzensen
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, United States
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Andrew Berchuck
- Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC, United States
| | - Malcolm C Pike
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States; Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Celeste Leigh Pearce
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, United States
| | - Penelope M Webb
- Gynaecological Cancers Group, Population Health Program, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia; University of Queensland, School of Public Health, Brisbane, Queensland, Australia.
| |
Collapse
|
29
|
Yip CSC. The associations of obesity with demographic and lifestyle factors among Hong Kong adults. Nutr Health 2025:2601060241303630. [PMID: 39901790 DOI: 10.1177/02601060241303630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Background: Obesity is a risk factor for leading causes of death. Aim: This study investigated the associations of obesity with demographic and lifestyle factors among adults aged 18-64 in Hong Kong. Methods: The study uses data (n = 24,855) from the government online database. It uses T-tests to compare mean values for body mass index (BMI), waist circumference, and weight-to-height ratio obese; analysis of variance for not-obese, BMI-obese-only, central-obese, and BMI + central-obese; Pearson chi-square tests for categorical variables; and multinomial logistic regression to obtain the odd ratios. It uses IBM SPSS version 26 to conduct all analyses, and at a 95% confidence level. Results: The analyses show that the risks of central-obese, and BMI + central-obese increase by 4%, and 4%/year of age, respectively. Females have 60%, 38%, and 60% lower risks of BMI-obese-only, central-obese, and BMI + central-obese, respectively, and people with tertiary education have 28%, 20%, and 20% lower risks, respectively. Employed people have a 40% higher risk of BMI-obese-only when compared with the unemployed; students have a 46% lower risk of BMI + central-obese and home-makers have a 38% higher risk. The risk of central-obese decreases by 14%/10 min/day of moderate physical activity, but could be different among females, and vigorous physical activity yielded mixed results. The risk of BMI + central-obese decreases by 9%/day/week of vegetable intake. Conclusions: Obesity is associated with multiple demographic and lifestyle factors. Keep doing vigorous and moderate physical activity but state alert to obesity risk factors, and frequent vegetable intake are recommended. Lifelong learning and continuing education could be an effective strategy to combat obesity.
Collapse
|
30
|
Qiu Y, Cao N, Meng D, Yuan J, Zhu Y. Survival and risk factors for metastatic colorectal cancer patients with a history of prior malignancy. Sci Rep 2025; 15:4045. [PMID: 39900612 PMCID: PMC11790972 DOI: 10.1038/s41598-025-88555-7] [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: 09/04/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025] Open
Abstract
Given concerns about treatment, there is uncertainty surrounding the effect of prior malignancy on the survival of individuals with metastatic colorectal cancer. This study sought to evaluate how prior malignancy impacts the survival of patients with metastatic colorectal cancer (mCRC). Patients diagnosed with stage IV mCRC (per the American Joint Committee on Cancer [AJCC] 6th edition) between 2004 and 2015 were identified from the Surveillance, Epidemiology, and End Results (SEER) database. Those without a prior history of malignancy were assigned to the control group, whereas those with a prior history of malignancy were assigned to the research group. Propensity score matching (PSM) was utilized to ensure that the baseline characteristics were balanced. The Kaplan‒Meier method was used for survival analysis, as were the multivariate Cox proportional hazard model and multivariate competing risk model. The PSM analysis included 54365 eligible patients with mCRC. Among them, 4,845 (8.9%) had a history of prior malignancy. A history of prior malignancy was associated with a greater cancer-specific survival rate (adjusted hazard ratio (AHR) ) = 0.49; 95% CI [0.47-0.51]). Subgroup analyses revealed that a prior diagnosis of a skin tumour (AHR = 1.37; 95% CI [1.11-1.69]) and a history of prior malignancy of more than five years (AHR = 1.39; 95% CI [1.23-1.57]) had adverse effects on the clinical outcomes of patients with mCRC. Our findings suggest that patients with a prior malignancy diagnosis may experience prolonged survival. Subgroup analysis indicated that a malignancy diagnosed more than 5 years ago may adversely impact the clinical outcomes of patients with mCRC. Therefore, we advocate for active standardized treatment for these patients and propose expanding the range of prior malignancies included in clinical trials based on publication timelines, primary tumour locations, and genetic testing results. The objective is to facilitate timely and proactive treatment for patients following the disclosure of results, thereby instilling confidence in the management of mCRC.
Collapse
Affiliation(s)
- Yiwen Qiu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Nida Cao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Dan Meng
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Jian Yuan
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Yingjie Zhu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
31
|
Hyun HK, Park JS, Park J, Park SJ, Park JJ, Cheon JH, Kim TI. Influence of Lifestyles on Polyp Burden and Cancer Development in Hereditary Colorectal Cancer Syndromes. J Gastroenterol Hepatol 2025; 40:433-445. [PMID: 39582265 DOI: 10.1111/jgh.16833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/14/2024] [Accepted: 11/09/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND Whether the progression of precursor lesions or the occurrence of cancer is influenced by lifestyle factors in carriers of genetic mutations has not been fully investigated, especially in Asian patients of hereditary colorectal cancer (CRC) syndrome. METHODS Patients at a high risk of hereditary CRC were included. For polyposis CRC syndromes, colorectal polyp burden was measured using at least 60 images per colonoscopy in each patient and classified into five stages using the International Society for Gastrointestinal Hereditary Tumours staging system according to the polyp number and size. Increase in tumor burden stage for polyposis CRC syndrome and the occurrence of CRC or any cancer for Lynch syndrome were analyzed according to lifestyle factors. RESULTS Ninety-six patients with suspected hereditary polyposis CRC syndrome and 106 patients with Lynch syndrome were recruited. For polyposis CRC syndromes, multivariate analysis showed that exposure to smoking and > 100 polyps independently predicted a high risk of increased polyp burden (p = 0.008 and p = 0.012, respectively). Significant genetic mutations or phenotype of polyposis syndromes were significantly associated with an increased polyp burden. For Lynch syndrome, smokers showed to be diagnosed with CRC in younger age than never-smokers (42.2 years vs. 49.0 years; p = 0.021), and heavy drinkers had high risk for occurrence of CRC (HR, 2.381, 95% CI, 1.338-4.236; p = 0.003) and any cancer (HR, 2.254; 95% CI, 1.334-3.806; p = 0.002). CONCLUSIONS The lifestyle factors (smoking and alcohol consumption) were associated with increasing precursor lesions and occurrence of cancer in patients with hereditary CRC syndrome. Lifestyle modifications may reduce the risk of hereditary CRC in carriers.
Collapse
Affiliation(s)
- Hye Kyung Hyun
- Department of Gastroenterology, Department of Internal Medicine, Yongin Severance Hospital, Yongin, Republic of Korea
| | - Ji Soo Park
- Hereditary Cancer Clinic, Cancer Prevention Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jihye Park
- Division of Gastroenterology, Department of Internal Medicine, and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo Jung Park
- Division of Gastroenterology, Department of Internal Medicine, and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Jun Park
- Hereditary Cancer Clinic, Cancer Prevention Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Gastroenterology, Department of Internal Medicine, and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae Hee Cheon
- Division of Gastroenterology, Department of Internal Medicine, and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae Il Kim
- Hereditary Cancer Clinic, Cancer Prevention Center, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
- Division of Gastroenterology, Department of Internal Medicine, and Institute of Gastroenterology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
Matsushita M, Takagi D, Kamada M. Socioeconomic Status and Patterns of Physical Activity and Sedentary Behavior: A Cross-Sectional Analysis of the J-SHINE. J Phys Act Health 2025; 22:262-269. [PMID: 39657642 DOI: 10.1123/jpah.2023-0138] [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: 03/29/2023] [Revised: 05/10/2024] [Accepted: 09/20/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND This study aimed to examine the association between socioeconomic status (SES) and optimal movement behaviors including sufficient moderate-to-vigorous physical activity (PA), regular leisure-time PA, and less sedentary behavior. METHODS We used data from the Japanese Study on Stratification, Health, Income, and Neighborhood conducted in 2012. Activity patterns and SES were measured using International Physical Activity Questionnaire Short-Version and other self-administered questionnaires. Activity pattern outcomes assessed were sufficient total volume of moderate-to-vigorous PA (≥150 min/wk), regular leisure-time PA (≥1 d/wk), less sedentary time (≤4 h/d), and optimal movement behaviors (meeting all criteria). SES variables were equivalized annual income, educational attainment, and occupation. Logistic regression analysis adjusted for confounders. RESULTS We included 731 men and 852 women. Lower income levels were associated with less leisure-time PA among men and women (men, odds ratio [OR]: 0.60 and 0.65 for Q2 and Q1 [lowest], respectively; women, OR: 0.53 and 0.56 for Q2 and Q1 [lowest]). Lower SES groups associated with less sedentary time (men, OR: 1.82 for blue-collar; women, OR: 1.44 for Q1 income level, 1.61 and 1.53 for junior college, technical school, and high school/junior high school, respectively; 1.89 for unemployed). SES was not significantly associated with optimal movement behavior in both sexes (in men, OR for those with high school or junior high school education vs those with university or graduate school education = 0.81 [0.43-1.53]). CONCLUSION Activity patterns varied by SES, with lower income associated with less leisure-time PA and white-collar workers associated with more prevalent sedentary behavior among both sexes.
Collapse
Affiliation(s)
- Munehiro Matsushita
- Department of Physical Recreation, School of Physical Education, Tokai University, Kanagawa, Japan
| | - Daisuke Takagi
- Department of Health and Social Behavior, School of Public Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masamitsu Kamada
- Department of Health Education and Health Sociology, School of Public Health, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
33
|
Zarean E, Li S, Wong EM, Makalic E, Milne RL, Giles GG, McLean C, Southey MC, Dugué PA. Evaluation of agreement between common clustering strategies for DNA methylation-based subtyping of breast tumours. Epigenomics 2025; 17:105-114. [PMID: 39711216 PMCID: PMC11792870 DOI: 10.1080/17501911.2024.2441653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024] Open
Abstract
AIMS Clustering algorithms have been widely applied to tumor DNA methylation datasets to define methylation-based cancer subtypes. This study aimed to evaluate the agreement between subtypes obtained from common clustering strategies. MATERIALS & METHODS We used tumor DNA methylation data from 409 women with breast cancer from the Melbourne Collaborative Cohort Study (MCCS) and 781 breast tumors from The Cancer Genome Atlas (TCGA). Agreement was assessed using the adjusted Rand index for various combinations of number of CpGs, number of clusters and clustering algorithms (hierarchical, K-means, partitioning around medoids, and recursively partitioned mixture models). RESULTS Inconsistent agreement patterns were observed for between-algorithm and within-algorithm comparisons, with generally poor to moderate agreement (ARI <0.7). Results were qualitatively similar in the MCCS and TCGA, showing better agreement for moderate number of CpGs and fewer clusters (K = 2). Restricting the analysis to CpGs that were differentially-methylated between tumor and normal tissue did not result in higher agreement. CONCLUSION Our study highlights that common clustering strategies involving an arbitrary choice of algorithm, number of clusters and number of methylation sites are likely to identify different DNA methylation-based breast tumor subtypes.
Collapse
Affiliation(s)
- Elaheh Zarean
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Shuai Li
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ee Ming Wong
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
| | - Enes Makalic
- Department of Data Science and AI, Faculty of Information Technology, Monash University, Clayton, VIC, Australia
| | - Roger L. Milne
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Graham G. Giles
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Catriona McLean
- Anatomical Pathology, Alfred Health, The Alfred Hospital, Melbourne, VIC, Australia
| | - Melissa C. Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Pierre-Antoine Dugué
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| |
Collapse
|
34
|
Miller KM, Liu C, Zhou Q, Iasonos A, Baser R, Ramesh B, Sonoda Y, Mueller JJ, Broach V, Abu-Rustum NR, Leitao MM. RELEVANT-C study: patient-reported prevalence of lower extremity lymphedema after sentinel lymph node mapping vs lymphadenectomy after surgery for early-stage cervical cancer. Int J Gynecol Cancer 2025; 35:100063. [PMID: 39971445 DOI: 10.1016/j.ijgc.2024.100063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVE To compare the prevalence of patient-reported lower extremity lymphedema and evaluate patient-reported quality of life after sentinel lymph node mapping vs comprehensive lymphadenectomy with or without sentinel lymph node mapping for the surgical management of early-stage cervical cancer. METHODS In July 2022, we mailed a survey that included a validated 13-item lower extremity lymphedema screening questionnaire to patients who underwent lymph node evaluation at the time of primary surgery for the 2018 International Federation of Gynecology and Obstetrics stage IA1 to IIB cervical cancer between January 1, 2006, and January 31, 2019. We excluded patients diagnosed with lower extremity lymphedema prior to surgery and those who answered ≤6 survey items, and we carried out 2 group comparisons: sentinel lymph node mapping vs lymphadenectomy with or without sentinel lymph node mapping, and patients with lower extremity lymphedema vs patients without. RESULTS Of 459 potential participants, 90 (20%) responded to the survey, all of which were evaluable (37 sentinel lymph nodes; 53 lymphadenectomies ± sentinel lymph nodes). Self-reported lower extremity lymphedema prevalence was 10.8% (4/37) in the sentinel lymph node mapping group and 43.4% (23/53) in the lymphadenectomy with or without sentinel lymph node mapping group (OR 6.32, 95% CI 2.14 to 23.5, p = .002). Histologic subtype and number of lymph nodes removed were associated with increased prevalence of lower extremity lymphedema. After adjusting for the histology subtype, lymphadenectomy retained independent association with an increased prevalence of lower extremity lymphedema over sentinel lymph node mapping (OR 4.96, 95% CI 1.61 to 18.8, p = .009). Patients with self-reported lower extremity lymphedema had significantly worse quality of life compared to those without self-reported lower extremity lymphedema. CONCLUSIONS We found sentinel lymph node mapping to be independently associated with a significantly decreased prevalence of patient-reported lower extremity lymphedema and with improved quality of life in patients undergoing surgical management of early-stage cervical cancer.
Collapse
Affiliation(s)
- Kathryn M Miller
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA
| | - Chrissy Liu
- SUNY Downstate Medical Center, Department of Obstetrics and Gynecology, New York, NY, USA
| | - Qin Zhou
- Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Alexia Iasonos
- Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Ray Baser
- Memorial Sloan Kettering Cancer Center, Department of Epidemiology and Biostatistics, New York, NY, USA
| | - Bhavani Ramesh
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA
| | - Yukio Sonoda
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA; Weill Cornell Medical College, Department of Obstetrics and Gynecology, New York, NY, USA
| | - Jennifer J Mueller
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA; Weill Cornell Medical College, Department of Obstetrics and Gynecology, New York, NY, USA
| | - Vance Broach
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA; Weill Cornell Medical College, Department of Obstetrics and Gynecology, New York, NY, USA
| | - Nadeem R Abu-Rustum
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA; Weill Cornell Medical College, Department of Obstetrics and Gynecology, New York, NY, USA
| | - Mario M Leitao
- Memorial Sloan Kettering Cancer Center, Department of Surgery, Gynecology Service, New York, NY, USA; Weill Cornell Medical College, Department of Obstetrics and Gynecology, New York, NY, USA.
| |
Collapse
|
35
|
Versluis MAJ, van de Poll-Franse LV, Zijlstra M, van Laarhoven HWM, Vreugdenhil G, Henselmans I, Brom L, Kuip EJM, van der Linden YM, Raijmakers NHJ. Changes in perception of prognosis in the last year of life of patients with advanced cancer and its associated factors: Longitudinal results of the eQuiPe study. Palliat Med 2025; 39:277-285. [PMID: 39629742 DOI: 10.1177/02692163241301220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
BACKGROUND Many patients with advanced cancer are unaware of their limited prognosis, however little is known about the change in awareness during the last year of their lives. AIM To investigate changes in the perception of prognosis in the last year of life of patients with advanced cancer and its associated factors. DESIGN Prospective, longitudinal, multicentre, observational study in patients with advanced cancer (eQuiPe). Patients completed 3-monthly follow-up questionnaires until death. SETTING/PARTICIPANTS Adult patients diagnosed with advanced cancer were recruited by their treating physician or self-enrolled in one of the forty Dutch hospitals. Only deceased patients with available prognostic data were included for analysis (n = 801). RESULTS Perception of prognosis changes in the last year of life with an increase in the percentage of patients who are aware of their limited prognosis (from 15% to 40%). Especially in the last 6 months of life, most of the changes were towards a more realistic perception of prognosis. Patients who did not want to know their prognosis remained relatively stable in their wish not to know (range: 14%-18%). Time to death was associated with having a perception of prognosis of < 1 year, >1 year or not knowing the prognosis, but was not associated with not wanting to know the prognosis. CONCLUSION Becoming aware of their limited prognosis may make patients with advanced cancer more receptive to start end-of-life discussions. Although some patients prefer not to know their prognosis, it remains important to respectfully explore their preferences and wishes for end-of-life care.
Collapse
Affiliation(s)
- M A J Versluis
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Graduate school of Social and behavioral sciences, Tilburg University, Tilburg, The Netherlands
| | - L V van de Poll-Franse
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Department of Medical and Clinical Psychology, Center for Research on Psychological and Somatic Diseases (CoRPS), Tilburg University, Tilburg, The Netherlands
- Department of Psychosocial Research and Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Zijlstra
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Department of Internal Medicine, St. Jans Gasthuis, Weert, The Netherlands
| | - H W M van Laarhoven
- Cancer Center Amsterdam, Cancer Treatment and Quality of Life, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam University Medical Centre location Amsterdam University, Amsterdam, The Netherlands
| | - G Vreugdenhil
- Department of Medical Oncology, Maxima Medical Centre, Veldhoven, The Netherlands
| | - I Henselmans
- Department of Medical Psychology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - L Brom
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| | - E J M Kuip
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Anaesthesiology, Pain and Palliative Care, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Y M van der Linden
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
- Centre of Expertise in Palliative Care, Leiden University Medical Centre, Leiden, the Netherlands
- Department of Radiotherapy, Leiden University Medical Centre, Leiden, the Netherlands
| | - N H J Raijmakers
- Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, The Netherlands
| |
Collapse
|
36
|
Kastora SL, Pantiora E, Hong YH, Veeramani M, Azim HA, Chakrabarti R, Geisler J, Knoop A, Lambertini M, Linderholm B, Meattini I, Partridge AH, Vaz-Luis I, Vorburger D, Yongue G, Karakatsanis A, Valachis A. Safety of topical estrogen therapy during adjuvant endocrine treatment among patients with breast cancer: A meta-analysis based expert panel discussion. Cancer Treat Rev 2025; 133:102880. [PMID: 39854791 DOI: 10.1016/j.ctrv.2025.102880] [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: 08/03/2024] [Revised: 12/15/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025]
Abstract
IMPORTANCE Endocrine treatments, such as Tamoxifen (TAM) and/or Aromatase inhibitors (AI), are the adjuvant therapy of choice for hormone-receptor positive breast cancer. These agents are associated with menopausal symptoms, adversely affecting drug compliance. Topical estrogen (TE) has been proposed for symptom management, given its' local application and presumed reduced bioavailability, however its oncological safety remains uncertain. OBJECTIVE The present systematic review, meta-analysis and expert panel review aimed to evaluate the strength of the available evidence on the risk of recurrence and mortality when TE is utilised in congruence with TAM or AI treatment, among BC survivors. DATA SOURCES Six databases and two prospective registers, were interrogated from inception to January 3rd, 2024. Search terms were Breast cancer AND Hormone replacement therapy AND topical/vaginal oestrogen AND recurrence/mortality. STUDY SELECTION All study designs reporting the use vs. non-use of TE in breast cancer survivors receiving adjuvant endocrine treatment were included. Six observational studies were deemed eligible for inclusion. DATA EXTRACTION AND SYNTHESIS Sources of heterogeneity were explored using subgroup analysis by risk of bias, median follow-up period, node positivity and menopausal status. Trial sequential analysis was performed to quantify outcome reliability. A global expert panel was called to deliberate on the data, pinpoint areas of limited understanding, and determine the most important areas for future research. MAIN OUTCOMES AND MEASURES Risk ratio effect sizes (RR) and corresponding 95 % Confidence Intervals (CI) of breast cancer recurrence and mortality in survivors on endocrine treatment (TAM and/or AI) exposed to TE were reported. Expert panel appraisal of meta-analysis evidence with definition of current knowledge gaps and future research aims. RESULTS In 38 050 female patients receiving adjuvant endocrine treatment, of whom 1805 had been exposed to TE, TE exposure of those on AI, did not increase all-cause mortality (RR 0.99 [95 %CI 0.58, 1.69], I2 = 81 %, P = 0.96; moderate GRADE certainty). However, such exposure may convey an increased risk of recurrence (RR 2.51 [95 % CI 1.10, 5.72], I2 = 9 %, P = 0.03; low-GRADE certainty). Exposure to TE during TAM did not increase either recurrence risk or all-cause mortality. Clinical factors such as lymph node positivity at the time of diagnosis and menopausal status and follow-up time appeared to be significant confounders. CONCLUSIONS AND RELEVANCE The use of TE does not appear to increase either recurrence or mortality risk among BC survivors treated with TAM. An increased recurrence risk, without an increase in mortality, cannot be ruled out when TE is used during AI.
Collapse
Affiliation(s)
- Stavroula L Kastora
- EGA Institute for Women's Health, University College London, Room G15, 86-96 Chenies Mews, London, UK.
| | - Eirini Pantiora
- Department of Surgical Sciences, Faculty of Medicine, Uppsala University, Uppsala, Sweden; Section for Breast Surgery, Department of Surgery, Uppsala University Hospital (Akademiska), Uppsala, Sweden.
| | - Yong Hwa Hong
- School of Medicine, St George's University of London, London, UK.
| | | | | | - Rima Chakrabarti
- EGA Institute for Women's Health, University College London, Room G15, 86-96 Chenies Mews, London, UK.
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog, Norway & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Ann Knoop
- Department of Oncology, Rigshospitalet, Denmark.
| | - Matteo Lambertini
- Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; Department of Internal Medicine and Medical Specialties (DIMI), School of Medicine, University of Genoa, 16132 Genoa, Italy.
| | - Barbro Linderholm
- Institution of Clinical Sciences,Department of Oncology, Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden.
| | - Icro Meattini
- Department of Experimental and Clinical Biomedical Sciences "M. Serio", University of Florence, Florence, Italy; Breast Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| | | | - Ines Vaz-Luis
- Cancer Survivorship Group, INSERM Unit 981, Interdisciplinary Department for the Organization of Patient Pathways - DIOPP, Medical Oncology Department, Gustave Roussy, Villejuif, France.
| | - Denise Vorburger
- Cantonal Hospital Winterthur, Department for Women's Health, Breast Unit, Winterthur, Switzerland.
| | - Gabriella Yongue
- EGA Institute for Women's Health, University College London, Room G15, 86-96 Chenies Mews, London, UK.
| | - Andreas Karakatsanis
- Department of Surgical Sciences, Faculty of Medicine, Uppsala University, Uppsala, Sweden; Section for Breast Surgery, Department of Surgery, Uppsala University Hospital (Akademiska), Uppsala, Sweden.
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden.
| |
Collapse
|
37
|
Lee MK, Mitchell SA, Basch E, Mazza GL, Langlais BT, Thanarajasingam G, Ginos BF, Rogak L, Meek EA, Jansen J, Deal AM, Carr P, Blinder VS, Jonsson M, Mody GN, Mendoza TR, Bennett AV, Schrag D, Dueck AC. Identification of meaningful individual-level change thresholds for worsening on the patient-reported outcomes version of the common terminology criteria for adverse events (PRO-CTCAE®). Qual Life Res 2025; 34:495-507. [PMID: 39503942 DOI: 10.1007/s11136-024-03819-5] [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: 10/21/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND We derived meaningful individual-level change thresholds for worsening in selected patient-reported outcomes version of the common terminology criteria for adverse events (PRO-CTCAE®) items and their composite scores. METHODS We used two data sources, the PRO-TECT trial (Alliance AFT-39) that collected PRO-CTCAE data from adults with advanced cancer at 26 United States (U.S.) community oncology practices and the PRO-CTCAE validation study that collected PRO-CTCAE data from adults undergoing chemotherapy or radiation therapy at nine U.S. cancer centers or community oncology practices. Both studies administered selected PRO-CTCAE items and EORTC QLQ-C30 scales. Conceptually, relevant QLQ-C30 domains were used as anchors to estimate meaningful change thresholds for deterioration in corresponding PRO-CTCAE items and their composite scores. Items or composites with ǀρǀ ≥ 0.30 correlation with QLQ-C30 scales were included. Changes in PRO-CTCAE scores and composites were estimated for patients who met or exceeded a 10-point deterioration on the corresponding QLQ-C30 scale. Change scores were computed between baseline and the 3-month timepoint in PRO-TECT, and in the PRO-CTCAE validation study between baseline and a single follow-up visit that occurred between 1 and 7 weeks later. For each PRO-CTCAE item, change scores could range from - 4 to 4; for a composite, change scores could range from - 3 to 3. RESULTS Change scores in QLQ-C30 and PRO-CTCAE were available in 406 and 792 patients in PRO-TECT and the validation study, respectively. Across QLQ-C30 scales, the proportion of patients with a 10-point or greater worsening on QLQ-C30 ranged from 15 to 30% in the PRO-TECT data and 13% to 34% in the validation data. Across PRO-CTCAE items, anchor-based meaningful change estimates for deterioration ranged from 0.05 to 0.30 (mean 0.19) in the PRO-TECT data and from 0.19 to 0.53 (mean 0.36) in the validation data. For composites, they ranged from 0.06 to 0.27 (mean 0.17) in the PRO-TECT data and 0.22 to 0.51 (mean 0.37) in the validation data. CONCLUSION In both datasets, the minimal meaningful individual-level change threshold for worsening was one point for all items and composite scores. CLINICALTRIALS gov: NCT03249090 (AFT-39), NCT02158637 (MC1091).
Collapse
Affiliation(s)
- Minji K Lee
- Alliance Foundation Trials Statistics and Data Center, Mayo Clinic, 200 1st Ave SW, Rochester, MN, 55902, USA.
| | | | - Ethan Basch
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gina L Mazza
- Alliance Foundation Trials Statistics and Data Center, Mayo Clinic, Scottsdale, AZ, USA
| | - Blake T Langlais
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Brenda F Ginos
- Alliance Foundation Trials Statistics and Data Center, Mayo Clinic, Scottsdale, AZ, USA
| | - Lauren Rogak
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, USA
| | - Eric A Meek
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, USA
| | - Jennifer Jansen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Allison M Deal
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philip Carr
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Mattias Jonsson
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Gita N Mody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Antonia V Bennett
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Deborah Schrag
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amylou C Dueck
- Alliance Foundation Trials Statistics and Data Center, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
38
|
Karipidis K, Baaken D, Loney T, Blettner M, Mate R, Brzozek C, Elwood M, Narh C, Orsini N, Röösli M, Paulo MS, Lagorio S. The effect of exposure to radiofrequency fields on cancer risk in the general and working population: A systematic review of human observational studies - Part II: Less researched outcomes. ENVIRONMENT INTERNATIONAL 2025; 196:109274. [PMID: 39904670 DOI: 10.1016/j.envint.2025.109274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 01/09/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND In the framework of the World Health Organization assessment of health effects of exposure to radiofrequency electromagnetic fields (RF-EMF), we have conducted a systematic review of human observational studies on the association between exposure to RF-EMF and risk of neoplastic diseases. Due to the extremely large number of included exposure types/settings and neoplasm combinations, we decided to present the review findings in two separate papers. In the first one we addressed the most investigated exposure-outcome pairs (e.g. glioma, meningioma, acoustic neuroma in relation to mobile phone use, or risk childhood leukemia in relation to environmental exposure from fixed-site transmitters) (Karipidis et al., 2024). Here, we report on less researched neoplasms, which include lymphohematopoietic system tumours, thyroid cancer and oral cavity/pharynx cancer, in relation to wireless phone use, or occupational RF exposure. METHODS Eligibility criteria: We included cohort and case-control studies of neoplasia risks in relation to three types of exposure to RF-EMF: 1. exposure from wireless phone use; 2. environmental exposure from fixed-site transmitters; 3. occupational exposures. In the current paper, we focus on less researched neoplasms including leukaemia, non-Hodgkin's lymphoma and thyroid cancer in mobile phone users; lymphohematopoietic system tumours and oral cavity/pharynx cancer in exposed workers. We focussed on investigations of specific neoplasms in relation to specific exposure sources (termed exposure-outcome pair, abbreviated E-O pairs), noting that a single article may address multiple E-O pairs. INFORMATION SOURCES Eligible studies were identified by predefined literature searches through Medline, Embase, and EMF-Portal. Risk-of-bias (RoB) assessment: We used a tailored version of the Office of Health Assessment and Translation (OHAT) RoB tool to evaluate each study's internal validity. Then, the studies were classified into three tiers according to their overall potential for bias (low, moderate and high) in selected, predefined and relevant bias domains. DATA SYNTHESIS We synthesized the study results using random effects restricted maximum likelihood (REML) models. Evidence assessment: Confidence in evidence was assessed according to the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) approach. RESULTS We included 26 articles, which were published between 1988 and 2019, with participants from 10 countries, reporting on 143 different E-O pairs, including 65 different types of neoplasms. Of these, 19 E-O pairs satisfied the criteria for inclusion in quantitative syntheses of the evidence regarding the risks of leukaemia, non-Hodgkin's lymphoma or thyroid cancer in relation to mobile phone use, and the risks of lymphohematopoietic system tumours or oral cavity/pharynx cancer following occupational exposure to RF-EMF. RF-EMF exposure from mobile phones (ever or regular use vs no or non-regular use) was not associated with an increased risk of leukaemia [meta-estimate of the relative risk (mRR) = 0.99, 95 % CI 0.91-1.07, 4 studies), non-Hodgkin's lymphoma (mRR = 0.99, 95 % CI = 0.92-1.06, 5 studies), or thyroid cancer (mRR = 1.05, 95 % CI = 0.88-1.26, 3 studies). Long-term (10 + years) mobile phone use was also not associated with risk of leukaemia (mRR = 1.03, 95 % CI 0.85-1.24, 3 studies), non-Hodgkin lymphoma (mRR = 0.99, 95 % CI 0.86-1.15, 3 studies), or thyroid cancer (no pooled estimate given the small number of studies). There were not sufficient studies of any specific neoplasms to perform dose-response meta-analyses for either cumulative call time or cumulative number of calls; individual studies did not show statistically significant associations between lifetime intensity of mobile phone use and any specific neoplasm. Occupational RF-EMF exposure (exposed vs unexposed) was not associated with an increased risk of lymphohematopoietic system tumours (mRR = 1.03, 95 % CI = 0.87-1.28, 4 studies) or oral cavity/pharynx cancer (mRR = 0.68, 95 % CI 0.42-1.11, 3 studies). There were not sufficient studies of any specific neoplasms to perform meta-analysis on the intensity or duration of occupational RF-EMF exposure; individual studies did not show statistically significant associations with either of those exposure metrics and any specific neoplasms. The small number of studies, and of exposed cases in some instances, hampered the assessment of the statistical heterogeneity in findings across studies in the meta-analyses. Based on the summary risk of bias, most studies included in the quantitative evidence syntheses were classified at moderate risk of bias. The most critical issue was exposure information bias, especially for occupational studies where the exposure characterization was rated at high risk of bias for all included studies. Outcome information bias was an issue in mortality-based occupational cohort studies investigating non-rapidly fatal neoplasms. Further, the healthy subscriber effect, and (at a lesser extent) the healthy worker effect, were identified as plausible explanations of the decreased risks observed in some studies. The association of RF-EMF exposure from wireless phone use, or workplace equipment/devices, with other important neoplasms was reported by only one or two studies per tumour, so no quantitative evidence syntheses were conducted on these outcomes. It is noted that there were generally no statistically significant exposure-outcome associations for any combinations, independently of the exposure metric and level, with a few studies reporting decreased risks (especially for smoking-related cancers). There was only one study which assessed the effect of RF-EMF exposure from fixed-site transmitters on less researched neoplasms and it reported no statistically significant associations between exposure from base stations and risk of lymphomas overall, lymphoma subtypes, or chronic lymphatic leukaemia in adults. CONCLUSIONS For near field RF-EMF exposure to the head from mobile phones, there was low certainty of evidence that it does not increase the risk of leukaemia, non-Hodgkin's lymphoma or thyroid cancer. For occupational RF-EMF exposure, there was very low certainty of evidence that it does not increase the risk of lymphohematopoietic system tumours or oral cavity/pharynx cancer. There was not sufficient evidence to assess the effect of whole-body far-field RF-EMF exposure from fixed-site transmitters (broadcasting antennas or base stations), or the effect of RF-EMF from any source on any other important neoplasms. OTHER This project was commissioned and partially funded by the World Health Organization (WHO). Co-financing was provided by the New Zealand Ministry of Health; the Istituto Superiore di Sanità in its capacity as a WHO Collaborating Centre for Radiation and Health; and ARPANSA as a WHO Collaborating Centre for Radiation Protection. REGISTRATION PROSPERO CRD42021236798. Published protocol: [(Lagorio et al., 2021) DOI https://doi.org/10.1016/j.envint.2021.106828].
Collapse
Affiliation(s)
- Ken Karipidis
- Australian Radiation Protection and Nuclear Safety Agency (ARPANSA) Yallambie VIC Australia.
| | - Dan Baaken
- Competence Center for Electromagnetic Fields, Federal Office for Radiation Protection (BfS) Cottbus Germany; Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University of Mainz, Germany
| | - Tom Loney
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences Dubai Health Dubai United Arab Emirates
| | - Maria Blettner
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University of Mainz, Germany
| | - Rohan Mate
- Australian Radiation Protection and Nuclear Safety Agency (ARPANSA) Yallambie VIC Australia
| | - Chris Brzozek
- Australian Radiation Protection and Nuclear Safety Agency (ARPANSA) Yallambie VIC Australia
| | - Mark Elwood
- Epidemiology and Biostatistics, School of Population Health, University of Auckland, New Zealand
| | - Clement Narh
- Department of Epidemiology and Biostatistics, School of Public Health (Hohoe Campus), University of Health and Allied Sciences, Ho PMB31, Ghana
| | - Nicola Orsini
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Martin Röösli
- Swiss Tropical and Public Health Institute, Basel, Switzerland; University of Basel, Basel, Switzerland
| | - Marilia Silva Paulo
- NOVA National School of Public Health, Public Health Research Center, Comprehensive Health Research Center, CHRC, REAL, CCAL, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Susanna Lagorio
- Department of Oncology and Molecular Medicine, National Institute of Health (Istituto Superiore di Sanità), Rome, Italy
| |
Collapse
|
39
|
Aliev F, De Sa Nogueira D, Aston-Jones G, Dick DM. Genetic associations between orexin genes and phenotypes related to behavioral regulation in humans, including substance use. Mol Psychiatry 2025:10.1038/s41380-025-02895-4. [PMID: 39880903 DOI: 10.1038/s41380-025-02895-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/23/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
The hypothalamic neuropeptide system of orexin (hypocretin) neurons provides projections throughout the neuraxis and has been linked to sleep regulation, feeding and motivation for salient rewards including drugs of abuse. However, relatively little has been done to examine genes associated with orexin signaling and specific behavioral phenotypes in humans. Here, we tested for association of twenty-seven genes involved in orexin signaling with behavioral phenotypes in humans. We tested the full gene set, functional subsets, and individual genes involved in orexin signaling. Our primary phenotype of interest was Externalizing, a composite factor comprised of behaviors and disorders associated with reward-seeking, motivation, and behavioral regulation. We also tested for association with additional phenotypes that have been related to orexin regulation in model organism studies, including alcohol consumption, problematic alcohol use, daytime sleepiness, insomnia, cigarettes per day, smoking initiation, and body mass index. The composite set of 27 genes corresponding to orexin function was highly associated with Externalizing, as well as with alcohol consumption, insomnia, cigarettes per day, smoking initiation and BMI. In addition, all gene subsets (except the OXR2/HCRTR2 subset) were associated with Externalizing. BMI was significantly associated with all gene subsets. The "validated factors for PPOX/HCRT" and "PPOX/HCRT upregulation" gene subsets also were associated with alcohol consumption. Individually, 8 genes showed a strong association with Externalizing, 12 with BMI, 7 with smoking initiation, 3 with alcohol consumption, and 2 with problematic alcohol use, after correction for multiple testing. This study indicates that orexin genes are associated with multiple behaviors and disorders related to self-regulation in humans. This is consistent with prior work in animals that implicated orexin signaling in motivational activation induced by salient stimuli, and supports the hypothesis that orexin signaling is an important potential therapeutic target for numerous behavioral disorders.
Collapse
Affiliation(s)
- Fazil Aliev
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - David De Sa Nogueira
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Gary Aston-Jones
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA
| | - Danielle M Dick
- Department of Psychiatry, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
- Rutgers Addiction Research Center, Brain Health Institute, Rutgers University and Rutgers Health, Piscataway, NJ, 08854, USA.
| |
Collapse
|
40
|
Xia Y, Guan X, Shi Z, Luo Y, He P. Remote symptom monitoring with patient-reported outcomes and nudges during lung cancer immunotherapy in China (PRO-NET): protocol for a randomised controlled trial. BMJ Open 2025; 15:e093374. [PMID: 39880457 PMCID: PMC11781134 DOI: 10.1136/bmjopen-2024-093374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025] Open
Abstract
INTRODUCTION Lung cancer is the leading cause of cancer-related mortality globally, with non-small cell lung cancer (NSCLC) comprising the majority of cases. For advanced NSCLC, immunotherapy offers substantial survival benefits but is often accompanied by severe immune-related adverse events symptoms, significantly affecting health-related quality of life (HRQoL). Routinely collection of patient-reported outcomes (PROs) followed by automated alerts has been shown to improve overall survival and HRQoL for cancers. However, there is limited evidence for PRO-based symptom monitoring on advanced NSCLC during immunotherapy. This study proposes an electronic PRO-based symptom tracking intervention, integrated with reactive alerts and nudges (PRO-NET) to improve HRQoL for advanced NSCLC patients receiving immunotherapy in China. Secondary objectives include assessing the effect of PRO-symptom monitoring on survival, physical function, symptom control, mental health, cost-effectiveness and implementation fidelity. METHODS AND ANALYSIS The PRO-NET study is a two-arm, parallel randomised controlled trial. The study will enrol at least 300 advanced NSCLC patients undergoing immunotherapy in China. Participants will be randomly assigned to either the intervention or control group in a ratio of 1:1 via PRO-NET programme. The intervention involves weekly electronic collection of immune-related PROs and reactive alerts sent directly to patients, combined with nudges over a 6-month period. Patients in the control group will follow usual care and will not trigger the alerts. Both the intervention and control groups will receive outcome assessments at baseline, 3 months and 6 months. Primary outcome focuses on HRQoL, while secondary outcomes include survival, physical function, symptom burden, mental health, cost-effectiveness and implementation fidelity. Differences in HRQoL between the groups will be compared using general linear mixed model, accounting for potential confounding. ETHICS AND DISSEMINATION The study was approved by the Institutional Review Board of the Peking University protocol on 21 July 2024 (No. IRB 00001052-24066). This protocol is based on V2.0, 6 July 2024 of the protocol. The results of this study will be disseminated through peer-reviewed publications and academic conferences. TRIALS REGISTRATION NUMBER ChiCTR2400088408.
Collapse
Affiliation(s)
- Yiqi Xia
- School of Public Health, Peking University, Beijing, China
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Xiaolong Guan
- School of Public Health, Peking University, Beijing, China
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Zhenyu Shi
- China Center for Health Development Studies, Peking University, Beijing, China
| | - Yanan Luo
- Department of Global Health, Peking University, Beijing, China
| | - Ping He
- China Center for Health Development Studies, Peking University, Beijing, China
| |
Collapse
|
41
|
Parvatam S, Pistollato F, Marshall LJ, Furtmann F, Jahagirdar D, Chakraborty Choudhury M, Mohanty S, Mittal H, Meganathan S, Mishra R. Human-based complex in vitro models: their promise and potential for rare disease therapeutics. Front Cell Dev Biol 2025; 13:1526306. [PMID: 39931243 PMCID: PMC11807990 DOI: 10.3389/fcell.2025.1526306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/09/2025] [Indexed: 02/13/2025] Open
Abstract
Rare diseases affect a small percentage of an individual country's population; however, with over 7,000 in total, rare diseases represent a significant disease burden impacting up to 10% of the world's population. Despite this, there are no approved treatments for almost 95% of rare diseases, and the existing treatments are cost-intensive for the patients. More than 70% of rare diseases are genetic in nature, with patient-specific mutations. This calls for the need to have personalised and patient-specific preclinical models that can lead to effective, speedy, and affordable therapeutic options. Complex in vitro models (CIVMs), including those using induced pluripotent stem cells (iPSCs), organoids, and organs-on-chips are emerging as powerful human-based pre-clinical systems with the capacity to provide efficacy data enabling drugs to move into clinical trials. In this narrative review, we discuss how CIVMs are providing insights into biomedical research on rare diseases. We also discuss how these systems are being used in clinical trials to develop efficacy models for rare diseases. Finally, we propose recommendations on how human relevant CIVMs could be leveraged to increase translatability of basic, applied and nonclinical research outcomes in the field of rare disease therapeutics in developed as well as middle-and low-income countries.
Collapse
Affiliation(s)
- Surat Parvatam
- Department of Research and Toxicology, Humane Society International/India, Hyderabad, India
| | - Francesca Pistollato
- Department of Research and Toxicology, Humane Society International/Europe, Brussels, Belgium
| | - Lindsay J. Marshall
- Animal Research Issues, The Humane Society of the United States, Washington DC, DC, United States
| | - Fabia Furtmann
- Department of Research and Toxicology, Humane Society International/Europe, Brussels, Belgium
| | - Devashree Jahagirdar
- Department of Chemical Engineering, Indian Institute of Technology (IIT), Mumbai, Maharashtra, India
| | | | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Harshita Mittal
- Department of Research and Toxicology, Humane Society International/India, Hyderabad, India
| | - Saveetha Meganathan
- Community Engagement and Policy Stewardship, Tata Institute for Genetics and Society, Bangalore, India
| | - Rakesh Mishra
- Tata Institute for Genetics and Society, Bangalore, India
| |
Collapse
|
42
|
Bernabeu E, Chybowska AD, Kresovich JK, Suderman M, McCartney DL, Hillary RF, Corley J, Valdés-Hernández MDC, Maniega SM, Bastin ME, Wardlaw JM, Xu Z, Sandler DP, Campbell A, Harris SE, McIntosh AM, Taylor JA, Yousefi P, Cox SR, Evans KL, Robinson MR, Vallejos CA, Marioni RE. Blood-based epigenome-wide association study and prediction of alcohol consumption. Clin Epigenetics 2025; 17:14. [PMID: 39863868 PMCID: PMC11762500 DOI: 10.1186/s13148-025-01818-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Alcohol consumption is an important risk factor for multiple diseases. It is typically assessed via self-report, which is open to measurement error through recall bias. Instead, molecular data such as blood-based DNA methylation (DNAm) could be used to derive a more objective measure of alcohol consumption by incorporating information from cytosine-phosphate-guanine (CpG) sites known to be linked to the trait. Here, we explore the epigenetic architecture of self-reported weekly units of alcohol consumption in the Generation Scotland study. We first create a blood-based epigenetic score (EpiScore) of alcohol consumption using elastic net penalized linear regression. We explore the effect of pre-filtering for CpG features ahead of elastic net, as well as differential patterns by sex and by units consumed in the last week relative to an average week. The final EpiScore was trained on 16,717 individuals and tested in four external cohorts: the Lothian Birth Cohorts (LBC) of 1921 and 1936, the Sister Study, and the Avon Longitudinal Study of Parents and Children (total N across studies > 10,000). The maximum Pearson correlation between the EpiScore and self-reported alcohol consumption within cohort ranged from 0.41 to 0.53. In LBC1936, higher EpiScore levels had significant associations with poorer global brain imaging metrics, whereas self-reported alcohol consumption did not. Finally, we identified two novel CpG loci via a Bayesian penalized regression epigenome-wide association study of alcohol consumption. Together, these findings show how DNAm can objectively characterize patterns of alcohol consumption that associate with brain health, unlike self-reported estimates.
Collapse
Affiliation(s)
- Elena Bernabeu
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Aleksandra D Chybowska
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Jacob K Kresovich
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Matthew Suderman
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 1TH, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Janie Corley
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Maria Del C Valdés-Hernández
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
- Centre for Clinical Brain Sciences, Edinburgh Imaging and UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Susana Muñoz Maniega
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
- Centre for Clinical Brain Sciences, Edinburgh Imaging and UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Mark E Bastin
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
- Centre for Clinical Brain Sciences, Edinburgh Imaging and UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Joanna M Wardlaw
- Edinburgh Medical School, Usher Institute, University of Edinburgh, Edinburgh, UK
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
- Scottish Imaging Network, A Platform for Scientific Excellence (SINAPSE) Collaboration, Edinburgh, UK
- Centre for Clinical Brain Sciences, Edinburgh Imaging and UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- Neurovascular Imaging Research Core, UCLA, Los Angeles, CA, USA
| | - Zongli Xu
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Andrew M McIntosh
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, UK
| | - Jack A Taylor
- Neurovascular Imaging Research Core, UCLA, Los Angeles, CA, USA
| | - Paul Yousefi
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 1TH, UK
| | - Simon R Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | - Catalina A Vallejos
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
- The Alan Turing Institute, London, UK.
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
43
|
Gliwska E, Głąbska D, Zaczek Z, Sobocki J, Guzek D. Nutritional Status and Information Provided to Polish Cancer Patients Assessed Using the EORTC QLQ-INFO25 Questionnaire. J Clin Med 2025; 14:697. [PMID: 39941366 PMCID: PMC11818191 DOI: 10.3390/jcm14030697] [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: 12/03/2024] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Malnutrition in cancer patients may significantly affect various aspects of the quality of life, outcomes, and prognosis, while satisfaction with the information provided may also influence these aspects. This study aims to assess the nutritional status of Polish cancer patients and its association with the level of information received, their potential need for more information, and the resultant quality of life. Methods: A cross-sectional study was conducted in 104 cancer patients. Validated European Organization for Research and Treatment of Cancer questionnaires EORTC QLQ-C30 and EORTC QLQ-INFO25 were used, and nutritional assessment was conducted using Subjective Global Assessment (SGA). Results: Male patients reported receiving more information than females about the disease, treatment, and care options, as well as greater satisfaction, and a higher overall score. Patients receiving enteral nutrition were more satisfied compared to those not receiving it, even if the scores for the information obtained within the specific areas did not differ, but they still wished to receive more information. Older patients reported higher scores than younger patients, indicating a higher level of information received regarding medical tests and higher satisfaction. The EORTC QLQ-INFO25 global score showed strong or moderate positive correlations with the majority of modules, and the level of information provided significantly influenced satisfaction. Conclusions: Female patients, those not receiving enteral nutrition, and young patients were less satisfied with the information received, which may negatively influence their quality of life. Effective communication with patients highlights the need for personalized informational support to enhance quality of life.
Collapse
Affiliation(s)
- Elwira Gliwska
- Department of Food Market and Consumer Research, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159C Nowoursynowska Street, 02-776 Warsaw, Poland;
- Cancer Epidemiology and Primary Prevention Department, Maria Sklodowska-Curie National Research Institute of Oncology, 15B Wawelska Street, 02-034 Warsaw, Poland
| | - Dominika Głąbska
- Department of Dietetics, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159C Nowoursynowska Street, 02-776 Warsaw, Poland;
| | - Zuzanna Zaczek
- Department of Human Nutrition, Faculty of Health Sciences, Medical University of Warsaw, 27 Erazma Ciolka Street, 01-445 Warsaw, Poland;
- Department of General Surgery and Clinical Nutrition, Centre of Postgraduate Medical Education in Warsaw, 231 Czerniakowska Street, 00-416 Warsaw, Poland;
| | - Jacek Sobocki
- Department of General Surgery and Clinical Nutrition, Centre of Postgraduate Medical Education in Warsaw, 231 Czerniakowska Street, 00-416 Warsaw, Poland;
| | - Dominika Guzek
- Department of Food Market and Consumer Research, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences (WULS-SGGW), 159C Nowoursynowska Street, 02-776 Warsaw, Poland;
| |
Collapse
|
44
|
Chen H, Chang SJ, Zheng Y, Zhou Y, Sun B, Hua C, Lin X. Analysis of Research Waste in Phase III/IV Randomized Controlled Trials of Botulinum Toxin Type A. Dermatol Surg 2025:00042728-990000000-01094. [PMID: 39840849 DOI: 10.1097/dss.0000000000004552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BACKGROUND The application of randomized controlled trials (RCTs) outcomes of botulinum toxin type A (BoNT-A) to actual patient care faces a significant hurdle due to research waste (RW). This includes the results of RCTs that have not been published even 4 years after completion and those with insufficiently reported outcomes and design flaws. The prevalence of RW within BoNT-A-associated RCTs remains uncertain. OBJECTIVE To assess the extent of RW (non-publication, insufficient reporting, and design flaws) in BoNT-A RCTs. METHODS The authors searched the ClinicalTrials using 'botulinum toxin type A' as the keyword. They verified the publication status through PubMed and Scopus searches. The adequacy of reporting was assessed using the Consolidated Standards of Reporting Trials checklist, while design limitations were evaluated based on the risk of bias and systematic reviews. RESULTS Two hundred twenty-eight RCTs met the inclusion criteria for characteristics presentation. The RW analysis omitted 53 RCTs completed after June 2020, which remained unpublished. Of the 175 RCTs assessed, 98 were published, 71 demonstrated adequate reporting, and 25 had design limitations. Altogether, 118 RCTs (67.4%) exhibited at least 1 trait of RW. Multicenter designs and registrations post-2014 were independent protective factors against RW. CONCLUSION Within the 175 RCTs included in RW analysis, 67.4% of the studies presented some form of RW. The varied characteristics of the identified RW indicators offer essential insights for more rational planning of future BoNT-A RCTs.
Collapse
Affiliation(s)
- Hongrui Chen
- All authors are affiliated with the Department of Plastic & Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Li JS, Riggins K, Yang L, Chen C, Castro P, Alfarkh W, Zarrin-Khameh N, Scheurer ME, Creighton CJ, Musher B, Li W, Shen L. DNA methylation profiling at base-pair resolution reveals unique epigenetic features of early-onset colorectal cancer in underrepresented populations. Clin Epigenetics 2025; 17:11. [PMID: 39844333 PMCID: PMC11753045 DOI: 10.1186/s13148-025-01817-z] [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: 07/13/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND The incidence of early-onset colorectal cancer (EOCRC) has been rising at an alarming rate in the USA, and EOCRC disproportionately affects racial/ethnic minorities. Here, we construct comprehensive profiles of EOCRC DNA methylomes at base-pair resolution for a cohort of Hispanic and African American patients. RESULTS We show the epigenetic landscape of these EOCRC patients differs from that of late-onset colorectal cancer patients, and methylation canyons in EOCRC tumor tissue preferentially overlapped genes in cancer-related pathways. Furthermore, we identify epigenetic alterations in metabolic genes that are specific to our racial/ethnic minority EOCRC cohort but not Caucasian patients from TCGA. Top genes differentially methylated between these cohorts included the obesity-protective MFAP2 gene as well as cancer risk susceptibility genes APOL3 and RNASEL. CONCLUSIONS In this study, we provide to the scientific community high-resolution DNA methylomes for a cohort of EOCRC patients from underrepresented populations. Our exploratory findings in this cohort highlight epigenetic mechanisms underlying the pathogenesis of EOCRC and nominate novel biomarkers for EOCRC in underrepresented populations.
Collapse
Affiliation(s)
- Jason Sheng Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Karen Riggins
- Department of Medicine, Hematology and Oncology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Li Yang
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chaorong Chen
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Patricia Castro
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Wedad Alfarkh
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Neda Zarrin-Khameh
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Pathology, Ben Taub Hospital, 1504 Taub Loop, Houston, TX, 77030, USA
| | - Michael E Scheurer
- Department of Pediatrics, Center for Epidemiology and Population Health, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chad J Creighton
- Department of Medicine and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin Musher
- Department of Medicine, Gastrointestinal Medical Oncology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Wei Li
- Division of Computational Biomedicine, Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA, 92697, USA.
| | - Lanlan Shen
- Department of Pediatrics, USDA Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
46
|
Galouzis N, Khawam M, Alexander EV, Mesropyan L, Luu C, Khreiss MR, Riall TS. Quality of life and social health in patients after pancreatic surgery. J Gastrointest Surg 2025; 29:101969. [PMID: 39848313 DOI: 10.1016/j.gassur.2025.101969] [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: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Clinicians lack robust data on quality of life (QOL) and social functioning after pancreatectomy limiting their ability guide patient decision making aligned with patients' goals of care. METHODS In this cross-sectional survey study, we administered the European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire, pancreas-specific QLQ-PAN26, Patient-Reported Outcomes Measurement Information System (PROMIS) Ability to Participate in Social Roles, and PROMIS activities and social isolation scales to all elective pancreatectomies (2021-2023). Results were compared with both normative data and between groups to determine factors predicting better QOL with a >10-12-point change considered clinically significant. RESULTS A total of 143 patients were included; 71 (49.6%) completed the distributed surveys. The average age of responders was 59.9 ± 16.1 years with 56.3% men. Pancreaticoduodenectomy (54.9%) was performed for malignancy in 67.6% of cases. Compared with normative population controls, postpancreatectomy patients reported lower role functioning scores (67.2 ± 28.7 vs 81.7 ± 28.2, 14.5 score difference) but less social isolation (40.6 ± 5.7 vs 50.0 ± 10.0, 9.4 score difference). Compared with patients with benign disease, those with malignancy reported clinically significant worse social functioning; more fatigue, pain, constipation, change in taste, weight loss, weakness, and altered bowel habits; worse body image; and increased worries about the future. Despite more symptoms, they were more satisfied with the healthcare they received (all >10-point score difference). CONCLUSION QOL and social health are affected by pancreatic resection and outcomes differ whether surgery was performed for benign or malignant disease. These issues are largely unaddressed and are potential targets for intervention to improve QOL.
Collapse
Affiliation(s)
- Nicholas Galouzis
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Maria Khawam
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | | | - Lusine Mesropyan
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | - Carrie Luu
- Department of Surgery, University of Arizona, Tucson, AZ, United States
| | | | - Taylor S Riall
- Department of Surgery, University of Arizona, Tucson, AZ, United States.
| |
Collapse
|
47
|
Yan C, Ma J, Zheng X, Shu J, Wang X, Chen X, Gao F, Li X, M S, Huang L, Yu X, Chen X. Diagnostic accuracy of human papillomavirus testing on self-collected samples among women referred for colposcopy. Sci Rep 2025; 15:2513. [PMID: 39833459 PMCID: PMC11747457 DOI: 10.1038/s41598-025-86943-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
To assess the diagnostic accuracy of self-collected urine and vaginal samples for the identification of precancerous cervical lesions in the referral population using high-risk human papillomavirus (hrHPV) assays based on polymerase chain reaction (PCR). It was a prospective study carried out in China from June 2021 to March 2022. The vaginal and urine samples were collected and analyzed by using a newly developed specific hrHPV PCR test, and matched cervical samples were analyzed by using an approved hrHPV DNA test. The primary outcomes were sensitivity for cervical intraepithelial neoplasia 2 or greater (CIN2 +). The secondary outcome was the accuracy of hrHPV findings in urine and vaginal samples compared to cervical samples. A total of 1,701 women were recruited with 113 women excluded. Among 1,588 qualified participants, a total of 203 cases of the CIN2 + group were enrolled in the two centers. The sensitivity and specificity of HPV detection for CIN2 + in urine, vaginal and cervical samples were 86.70% and 36.46%, 90.64% and 30.54%, 93.60% and 26.14%, respectively. The urine sample performed lower sensitivity (p = 0.003) and higher specificity (p < 0.001) than the cervical sample. There was no difference in sensitivity between vaginal and cervical samples (p = 0.146), and the specificity of vaginal samples was higher than that in cervical samples (p < 0.001). The agreement was 78.15% of urine and cervical samples and 85.71% of vaginal and cervical samples. HPV testing on self-collected urine and vaginal samples has acceptable consistency compared with traditional cervical samples. It may be an alternative option for cervical cancer screening. Additional studies are still required to substantiate this issue.
Collapse
Affiliation(s)
- ChunX Yan
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jiong Ma
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xia Zheng
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Shu
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - XiaoJ Wang
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - XiaoY Chen
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Feng Gao
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - XiaoQ Li
- Hangzhou New Horizon Health Technology Co., Ltd, Hangzhou, Zhejiang, China
| | - ShanQ M
- Hangzhou New Horizon Health Technology Co., Ltd, Hangzhou, Zhejiang, China
| | - LongM Huang
- Hangzhou New Horizon Health Technology Co., Ltd, Hangzhou, Zhejiang, China
| | - XiaoT Yu
- Hangzhou New Horizon Health Technology Co., Ltd, Hangzhou, Zhejiang, China
| | - XueJ Chen
- The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Llaha F, Licaj I, Sharashova E, Benjaminsen Borch K, Lukic M. Alcohol consumption trajectories and associated factors in adult women: the Norwegian Women and Cancer study. Alcohol Alcohol 2025; 60:agaf005. [PMID: 39921373 PMCID: PMC11806201 DOI: 10.1093/alcalc/agaf005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
AIMS We described the age-specific trajectories of total alcohol consumption and the consumption of different types of beverages among adult Norwegian women as they age, and how these relate to education, lifestyle, and health-related factors. METHODS This study included 76 382 women aged 31-70 years who participated in at least two of the three Norwegian Women and Cancer (NOWAC) study surveys conducted in 1991-97, 1998-2003, and 2004-11. Group-based trajectory modeling was used to identify the trajectories of self-reported alcohol consumption. Multinomial regression models were used to fit the adjusted odds ratios (ORs) of the associations between education, lifestyle, health-related factors, and the trajectory membership. Analysis was stratified into two subcohorts: women aged 31-49 years and women aged 50-70 years at enrolment. RESULTS Five different trajectories of total alcohol consumption were identified among the two subcohorts: non-drinker stable (12.5%-23.6%), low stable (66.3%-60.1%), light increasing or light unstable (17.8%-12.1%), moderate to high or light to high (2.8%-2.7%), and high to moderate or moderate decreasing (.6%-1.4%). Trajectories were resembled by those of wine consumption. Compared to low stable drinkers, women who sustained or increased their total alcohol consumption showed higher ORs for higher education level, excellent self-rated health, former or current smoking status, and a body mass index (BMI) below 25 kg/m2. CONCLUSION While most women in this study maintained stable low-light levels of alcohol consumption, certain groups-such as women with higher education and better health-were more likely to increase their drinking with age. Women can particularly increase their drinking around the retirement age. The increasing trends of total alcohol consumption were reflected by those of wine. These findings provide information into groups and beverages that could be targeted in alcohol-reducing interventions.
Collapse
Affiliation(s)
- Fjorida Llaha
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Idlir Licaj
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | - Ekaterina Sharashova
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| | | | - Marko Lukic
- Department of Community Medicine, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
49
|
Zarean E, Li S, Wong EM, Makalic E, Milne RL, Giles GG, McLean C, Southey MC, Dugué PA. Tumour DNA methylation markers associated with breast cancer survival: a replication study. Breast Cancer Res 2025; 27:9. [PMID: 39825380 PMCID: PMC11740461 DOI: 10.1186/s13058-024-01955-x] [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: 09/23/2024] [Accepted: 12/20/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Tumour DNA methylation has been investigated as a potential marker for breast cancer survival, but findings often lack replication across studies. METHODS This study sought to replicate previously reported associations for individual CpG sites and multi-CpG signatures using an Australian sample of 425 women with breast cancer from the Melbourne Collaborative Cohort Study (MCCS). Candidate methylation sites (N = 22) and signatures (N = 3) potentially associated with breast cancer survival were identified from five prior studies that used The Cancer Genome Atlas (TCGA) methylation dataset, which shares key characteristics with the MCCS: comparable sample size, tissue type (formalin-fixed paraffin-embedded; FFPE), technology (Illumina HumanMethylation450 array), and participant characteristics (age, ancestry, and disease subtype and severity). Cox proportional hazard regression analyses were conducted to assess associations between these markers and both breast cancer-specific survival and overall survival, adjusting for relevant participant characteristics. RESULTS Our findings revealed partial replication for both individual CpG sites (9 out of 22) and multi-CpG signatures (2 out of 3). These associations were maintained after adjustment for participant characteristics and were stronger for breast cancer-specific mortality than for overall mortality. In fully-adjusted models, strong associations were observed for a CpG in PRAC2 (per standard deviation [SD], HR = 1.67, 95%CI: 1.24-2.25) and a signature based on 28 CpGs developed using elastic net (per SD, HR = 1.48, 95%CI: 1.09-2.00). CONCLUSIONS While further studies are needed to confirm and expand on these findings, our study suggests that DNA methylation markers hold promise for improving breast cancer prognostication.
Collapse
Affiliation(s)
- Elaheh Zarean
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Shuai Li
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
| | - Ee Ming Wong
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Enes Makalic
- Department of Data Science and AI, Faculty of Information Technology, Monash University, Clayton, VIC, Australia
| | - Roger L Milne
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Graham G Giles
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
| | - Catriona McLean
- Anatomical Pathology, Alfred Health, The Alfred Hospital, Melbourne, VIC, Australia
| | - Melissa C Southey
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia
- Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, VIC, Australia
| | - Pierre-Antoine Dugué
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia.
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, VIC, Australia.
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, VIC, Australia.
| |
Collapse
|
50
|
Loroña NC, Himbert C, Ose J, Cohen SA, Strehli I, Ulrich CM, Cobos S, Baptiste EJ, Bloomer AM, Figueiredo JC, Gigic B, Hardikar S, Karchi M, Mutch M, Peoples AR, Schneider M, Shibata D, Siegel EM, Toriola AT, Wood EH, Li CI. Alcohol Consumption and Smoking History at the Time of Diagnosis and the Risk of Colorectal Cancer Recurrence and Mortality: Results from the ColoCare Study. Cancer Epidemiol Biomarkers Prev 2025; 34:59-66. [PMID: 39373623 PMCID: PMC11717602 DOI: 10.1158/1055-9965.epi-24-0834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/07/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Findings from studies investigating the impacts of alcohol use and smoking on colorectal cancer outcomes are inconclusive. This study aimed to investigate associations between alcohol use and smoking status at the time of diagnosis on recurrence and overall mortality among patients with colorectal cancer. METHODS The present study included 2,216 stage I-IV patients with colorectal cancer from the longitudinal multicenter ColoCare Study, with available data on recurrence and colorectal cancer-specific mortality. Cox proportional hazards models adjusted for age, sex, race, ethnicity, stage, tumor site, treatment, comorbidities, body mass index, and study site were fit, with imputations for missing data. RESULTS We observed 235 recurrences and 308 colorectal cancer-specific deaths over an average of 3 years of follow-up. After adjusting for confounders, current alcohol consumption and ever smoking, relative to not current consumption and never smoking, respectively, were not statistically significantly associated with colorectal cancer recurrence [alcohol-HR, 0.95. 95% confidence interval (CI), 0.71-1.29; ever smoking-HR, 0.98, 95% CI, 0.75-1.29] or colorectal cancer-specific mortality (alcohol-HR, 0.95. 95% CI, 0.74-1.22; ever smoking-HR, 0.98, 95% CI, 0.77-1.24). CONCLUSIONS No associations were observed between alcohol and smoking at diagnosis and clinical outcomes in this well-annotated longitudinal cohort. IMPACT Our cohort study reports no significant associations; however, limiting alcohol use and avoiding smoking are health behaviors recommended for colorectal cancer survivors for prevention of other cancers and chronic conditions.
Collapse
Affiliation(s)
- Nicole C Loroña
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles CA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Caroline Himbert
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Jennifer Ose
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
- University of Applied Sciences and Arts, Department of Media, Information, and Design, Hannover, Germany
| | - Stacey A Cohen
- Division of Hematology/Oncology, University of Washington, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ildiko Strehli
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Cornelia M Ulrich
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Sofia Cobos
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Esther Jean Baptiste
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
- Non-Therapeutic Research Office, Moffitt Cancer Center, Tampa, FL, USA
| | - Amanda M Bloomer
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jane C Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles CA, USA
- Department of Computational Biomedicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Biljana Gigic
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Sheetal Hardikar
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Meghana Karchi
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Matthew Mutch
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, St. Louis, MO, USA
| | - Anita R Peoples
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Department of General, Visceral, Thoracic, Transplantation and Pediatric Surgery, Giessen University Hospital, Giessen, Germany
| | - David Shibata
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Erin M. Siegel
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
- Non-Therapeutic Research Office, Moffitt Cancer Center, Tampa, FL, USA
| | - Adetunji T Toriola
- Siteman Cancer Center, St. Louis, MO, USA
- Department of Surgery, Division of Public Health Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Christopher I Li
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|