201
|
Cagigas ML, De Ciutiis I, Masedunskas A, Fontana L. Dietary and pharmacological energy restriction and exercise for healthspan extension. Trends Endocrinol Metab 2025:S1043-2760(25)00076-1. [PMID: 40318928 DOI: 10.1016/j.tem.2025.04.001] [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: 02/23/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 05/07/2025]
Abstract
Extending healthspan - the years lived in optimal health - holds transformative potential to reduce chronic diseases and healthcare costs. Dietary restriction (DR), particularly when combined with nutrient-rich diets and exercise, is among the most effective, evidence-based strategies for enhancing metabolic health and longevity. By targeting fundamental pathways, it mitigates the onset and progression of obesity, type 2 diabetes (T2D), cardiovascular disease (CVD), neurodegeneration, and cancer. This review synthesizes human data on the impact of DR and exercise on metabolic and age-related diseases, while emphasizing key biological mechanisms such as nutrient sensing, insulin sensitivity, inflammation, mitochondrial function, and gut microbiota. We also examine the emerging role of pharmacologically induced DR, focusing on glucagon-like peptide 1 (GLP-1) receptor agonists (RAs) that partially mimic DR and present opportunities for chronic disease prevention.
Collapse
Affiliation(s)
- Maria Lastra Cagigas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Isabella De Ciutiis
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Andrius Masedunskas
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Luigi Fontana
- Charles Perkins Center, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia.
| |
Collapse
|
202
|
Lee SH, Song DS, Kim UC, Jee SH, Lee K. The Causal Relationship between Telomere Length and Cancer Risk: A Two-Sample Mendelian Randomization. Cancer Epidemiol Biomarkers Prev 2025; 34:737-743. [PMID: 40079752 PMCID: PMC12046325 DOI: 10.1158/1055-9965.epi-24-1168] [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: 08/11/2024] [Revised: 11/02/2024] [Accepted: 03/10/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Telomere length (TL) shortens with age and is associated with an increased risk of numerous chronic diseases. However, the causal direction of the association between TL and cancer risk remains uncertain. This study aimed to assess the causal impact of TL on cancer risk using Mendelian randomization (MR) analysis. METHODS Genome-wide association studies from Singapore and China data, the Korean Cancer Prevention Study II (KCPS-II), the Korean Genome Epidemiologic Study, and the Biobank of Japan were utilized. A two-sample MR study was performed using summary-level genome-wide association study data from individuals of East Asian ancestry. SNPs associated with TL were used as instrumental variables. RESULTS Longer TL per 1-SD increase due to germline genetic variants was associated with a higher risk of site-specific cancer. In the KCPS-II and Korean Genome Epidemiologic Study, the strongest association was observed with thyroid cancer {OR, 2.49 [95% confidence interval (CI), 1.79-3.47] and 2.27 (1.49-3.46)}, followed by lung cancer [OR, 2.19 (95% CI, 1.60-3.08) and 1.45 (1.12-1.87)]. Similar results were observed in the Biobank of Japan, with OR, 2.92 (95% CI, 1.75-4.88) for thyroid cancer and 2.04 (1.41-2.94) for lung cancer. In histologic subgroup analysis of KCPS-II, a significant relationship was found with lung adenocarcinoma [OR, 2.26 (95% CI, 1.55-3.31)] but not with lung squamous cell carcinoma (1.21, 0.47-3.06). After removing outlier SNPs in the radial MR analysis, significant associations were identified for both lung adenocarcinoma [OR, 1.88 (95% CI, 1.25-2.82)] and lung squamous cell carcinoma (2.29, 1.05-4.98). CONCLUSIONS Our findings suggest that longer TL increases the risk of various cancers in East Asian populations. IMPACT Genetically determined longer TL may contribute to a risk of certain cancers.
Collapse
Affiliation(s)
- Su Hyun Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, South Korea
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
- Department of Public Health, Graduate School, Yonsei University, Seoul, Korea
| | - Dae Sub Song
- Division of Population Health Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju-si, Republic of Korea
| | - Un Chong Kim
- Department of Biostatistics and Computing, College of Medicine, Yonsei University, Korea
| | - Sun Ha Jee
- Department of Epidemiology and Health Promotion, Institute for Health Promotion, Graduate School of Public Health, Yonsei University, Seoul, Korea
| | - Kyoungho Lee
- Division of Population Health Research, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju-si, Republic of Korea
| |
Collapse
|
203
|
Arenillas C, Celada L, Ruiz-Cantador J, Calsina B, Datta D, García-Galea E, Fasani R, Moreno-Cárdenas AB, Alba-Linares JJ, Miranda-Barrio B, Martínez-Montes ÁM, Alvarez-Escola C, Lecumberri B, González García A, K. Flores S, Esquivel E, Ding Y, Peitzsch M, Robles-Guirado JÁ, Regojo Zapata RM, Pozo-Kreilinger JJ, Iglesias C, Dwight T, Muir CA, Oleaga A, Garrido-Lestache Rodríguez-Monte ME, Del Cerro MJ, Martínez-Bendayán I, Álvarez-González E, Cubiella T, Lourenço DM, A. Pereira MA, Burnichon N, Buffet A, Broberg C, Dickson PV, Fraga MF, Llorente Pendás JL, Rueda Soriano J, Buendía Fuentes F, Toledo SP, Clifton-Bligh R, Dienstmann R, Villanueva J, Capdevila J, Gimenez-Roqueplo AP, Favier J, Nuciforo P, Young WF, Bechmann N, Opotowsky AR, Vaidya A, Bancos I, Weghorn D, Robledo M, Casteràs A, Dos-Subirà L, Adameyko I, Chiara MD, Dahia PL, Toledo RA. Convergent Genetic Adaptation in Human Tumors Developed Under Systemic Hypoxia and in Populations Living at High Altitudes. Cancer Discov 2025; 15:1037-1062. [PMID: 40199338 PMCID: PMC12046333 DOI: 10.1158/2159-8290.cd-24-0943] [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: 07/04/2024] [Revised: 11/21/2024] [Accepted: 01/27/2025] [Indexed: 04/10/2025]
Abstract
SIGNIFICANCE This study reveals a broad convergence in genetic adaptation to hypoxia between natural populations and tumors, suggesting that insights from natural populations could enhance our understanding of cancer biology and identify novel therapeutic targets. See related commentary by Lee, p. 875.
Collapse
Affiliation(s)
- Carlota Arenillas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Lucía Celada
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - José Ruiz-Cantador
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Debayan Datta
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Eduardo García-Galea
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Roberta Fasani
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ana Belén Moreno-Cárdenas
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Juan José Alba-Linares
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Berta Miranda-Barrio
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Ángel M. Martínez-Montes
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | | | - Beatriz Lecumberri
- Department of Endocrinology and Nutrition, Hospital Universitario La Paz, Madrid, Spain
| | - Ana González García
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitario La Paz, Madrid, Spain
| | - Shahida K. Flores
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Emmanuel Esquivel
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Yanli Ding
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Mirko Peitzsch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - José-Ángel Robles-Guirado
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | | - Carmela Iglesias
- Department of Pathology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Trisha Dwight
- Cancer Genetics, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
- The University of Sydney, Sydney, Australia
| | - Christopher A. Muir
- Department of Endocrinology, St. Vincent’s Hospital, Sydney, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Amelia Oleaga
- Department of Endocrinology and Nutrition, Hospital Universitario de Basurto, Bilbao, Spain
| | | | - Maria Jesús Del Cerro
- Department of Pediatric Cardiology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Isaac Martínez-Bendayán
- Department of Pediatric Cardiology, Instituto de Investigación Biomédica (Cardiopatía Estructural y Congénita) and Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Enol Álvarez-González
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Department of Functional Biology, Genetic Area, University of Oviedo, Oviedo, Spain
| | - Tamara Cubiella
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Delmar Muniz Lourenço
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Maria Adelaide A. Pereira
- Endocrinology Division, Hospital das Clínicas, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Nelly Burnichon
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Alexandre Buffet
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Craig Broberg
- Adult Congenital Heart Program, Division of Cardiology, Oregon Health and Science University, Portland, Oregon
| | - Paxton V. Dickson
- Department of Surgery, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Mario F. Fraga
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Cancer Epigenetics and Nanomedicine Laboratory, Nanomaterials and Nanotechnology Research Center (CINN-CSIC), University of Oviedo, Oviedo, Spain
- Department of Organisms and Systems Biology (B.O.S.), University of Oviedo, Oviedo, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Luis Llorente Pendás
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Joaquín Rueda Soriano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Francisco Buendía Fuentes
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Adult Congenital Heart Disease Unit, Department of Cardiology, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | | | - Roderick Clifton-Bligh
- Department of Endocrinology and Cancer Genetics Unit, Kolling Institute, Royal North Shore Hospital, Sydney, Australia
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- University of Vic – Central University of Catalonia, Vic, Spain
| | - Josep Villanueva
- Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jaume Capdevila
- Neuroendocrine and Endocrine Tumor Translational Research Program (NET-VHIO), Vall Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Department of Medical Oncology, Gastrointestinal and Endocrine Tumor Unit, Vall d’Hebron Hospital Universitari, Vall d’Hebron Hospital Campus, Barcelona, Spain
| | - Anne-Paule Gimenez-Roqueplo
- AP-HP, Hôpital Européen Georges Pompidou, Département de Médecine Génomique des Tumeurs et des Cancers, Paris, France
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Judith Favier
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | - Nicole Bechmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Alexander R. Opotowsky
- Cincinnati Adult Congenital Heart Disease Program, Heart Institute, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, Ohio
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Anand Vaidya
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Irina Bancos
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Anna Casteràs
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- Department of Endocrinology and Nutrition, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Laura Dos-Subirà
- Integrated Adult Congenital Heart Disease Unit, Department of Cardiology, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- European Reference Network for Rare, Low-Prevalence, or Complex Diseases of the Heart (ERN GUARD-Heart), Amsterdam, the Netherlands
| | - Igor Adameyko
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Department of Physiology and Pharmacology, Karolinska Institutet Stockholm, Sweden
| | - María-Dolores Chiara
- Health Research Institute of Asturias (ISPA), University of Oviedo, Oviedo, Spain
- Institute of Oncology of Asturias (IUOPA), University of Oviedo, Oviedo, Spain
| | - Patricia L.M. Dahia
- Division of Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center, San Antonio, Texas
| | - Rodrigo A. Toledo
- Biomarkers and Clonal Dynamics Group, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
204
|
Yamaguchi TN, Houlahan KE, Zhu H, Kurganovs N, Livingstone J, Fox NS, Yuan J, Sietsma Penington J, Jung CH, Schwarz T, Jaratlerdsiri W, van Riet J, Georgeson P, Mangiola S, Taraszka K, Lesurf R, Jiang J, Chow K, Heisler LE, Shiah YJ, Ramanand SG, Clarkson MJ, Nguyen A, Espiritu SMG, Stuchbery R, Jovelin R, Huang V, Bell C, O’Connor E, McCoy PJ, Lalansingh CM, Cmero M, Salcedo A, Chan EK, Liu LY, Stricker PD, Bhandari V, Bornman RM, Sendorek DH, Lonie A, Prokopec SD, Fraser M, Peters JS, Foucal A, Mutambirwa SB, Mcintosh L, Orain M, Wakefield M, Picard V, Park DJ, Hovington H, Kerger M, Bergeron A, Sabelnykova V, Seo JH, Pomerantz MM, Zaitlen N, Waszak SM, Gusev A, Lacombe L, Fradet Y, Ryan A, Kishan AU, Lolkema MP, Weischenfeldt J, Têtu B, Costello AJ, Hayes VM, Hung RJ, He HH, McPherson JD, Pasaniuc B, van der Kwast T, Papenfuss AT, Freedman ML, Pope BJ, Bristow RG, Mani RS, Corcoran NM, Reimand J, Hovens CM, Boutros PC. The Germline and Somatic Origins of Prostate Cancer Heterogeneity. Cancer Discov 2025; 15:988-1017. [PMID: 39945744 PMCID: PMC12046336 DOI: 10.1158/2159-8290.cd-23-0882] [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: 08/03/2023] [Revised: 12/06/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
SIGNIFICANCE This study uncovered 223 recurrently mutated driver regions using the largest cohort of prostate tumors to date. It reveals associations between germline SNPs, somatic drivers, and tumor aggression, offering significant insights into how prostate tumor evolution is shaped by germline factors and the timing of somatic mutations.
Collapse
Affiliation(s)
- Takafumi N. Yamaguchi
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
| | - Kathleen E. Houlahan
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Helen Zhu
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Natalie Kurganovs
- Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Julie Livingstone
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
| | - Natalie S. Fox
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jiapei Yuan
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | | | - Chol-Hee Jung
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California
| | - Weerachai Jaratlerdsiri
- Laboratory for Human Comparative and Prostate Cancer Genomics, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Job van Riet
- Department of Medical Oncology, Erasmus University, Rotterdam, the Netherlands
| | - Peter Georgeson
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Stefano Mangiola
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Kodi Taraszka
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California
| | - Robert Lesurf
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Jue Jiang
- Laboratory for Human Comparative and Prostate Cancer Genomics, Genomics and Epigenetics Theme, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Ken Chow
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
| | | | - Yu-Jia Shiah
- Ontario Institute for Cancer Research, Toronto, Canada
| | | | - Michael J. Clarkson
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Anne Nguyen
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Ryan Stuchbery
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Vincent Huang
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Connor Bell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edward O’Connor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick J. McCoy
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Marek Cmero
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Adriana Salcedo
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Eva K.F. Chan
- St Vincent’s Clinical School, University of New South Wales, Randwick, Australia
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
| | - Lydia Y. Liu
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
| | - Phillip D. Stricker
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
| | - Vinayak Bhandari
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Riana M.S. Bornman
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | | | - Andrew Lonie
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | | | - Michael Fraser
- Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Justin S. Peters
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Adrien Foucal
- Ontario Institute for Cancer Research, Toronto, Canada
| | | | - Lachlan Mcintosh
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Michèle Orain
- Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Matthew Wakefield
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Valérie Picard
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Daniel J. Park
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Hélène Hovington
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Michael Kerger
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
| | - Alain Bergeron
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | | | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M. Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Noah Zaitlen
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, California
| | - Sebastian M. Waszak
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Genetics, Brigham Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- The Eli and Edythe L. Broad Institute, Cambridge, Massachusetts
| | - Louis Lacombe
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Yves Fradet
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Andrew Ryan
- TissuPath Specialist Pathology Services, Mount Waverley, Australia
| | - Amar U. Kishan
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, California
| | - Martijn P. Lolkema
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California
- Center for Personalized Cancer Treatment, Rotterdam, the Netherlands
| | - Joachim Weischenfeldt
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bernard Têtu
- Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Anthony J. Costello
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
| | - Vanessa M. Hayes
- St Vincent’s Clinical School, University of New South Wales, Randwick, Australia
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
- Central Clinical School, University of Sydney, Camperdown, Australia
- Department of Medical Sciences, University of Limpopo, Mankweng, South Africa
| | - Rayjean J. Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Toronto, Canada
- Epidemiology Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Housheng H. He
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - John D. McPherson
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Bogdan Pasaniuc
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, California
| | | | - Anthony T. Papenfuss
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Department of Mathematics and Statistics, University of Melbourne, Parkville, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bernard J. Pope
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Medicine, Monash University, Clayton, Australia
| | - Robert G. Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Manchester Cancer Research Centre, Manchester, United Kingdom
| | - Ram S. Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Niall M. Corcoran
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
- Department of Urology, Peninsula Health, Frankston, Australia
- The Victorian Comprehensive Cancer Centre, Parkville, Australia
| | - Jüri Reimand
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Christopher M. Hovens
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
205
|
Tirkes T, Yadav D, Conwell DL, Zhao X, Dasyam AK, Halappa VG, Patel A, Shah ZK, Swensson J, Takahashi N, Venkatesh S, Wachsman A, Li L, Jennings K, Yang Y, Hart PA, Pandol SJ, Park WG, Vege SS, Topazian M, Territo PR, Persohn SA, Andersen DK, Fogel EL, the Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer (CPDPC). Multiparametric MRI Scoring System of the Pancreas for the Diagnosis of Chronic Pancreatitis. J Magn Reson Imaging 2025; 61:2183-2194. [PMID: 39225586 PMCID: PMC11873175 DOI: 10.1002/jmri.29594] [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: 02/12/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Ductal features alone may not offer high diagnostic sensitivity or most accurate disease severity of chronic pancreatitis (CP). PURPOSE Diagnose CP based on multiparametric MRI and MRCP features. STUDY TYPE Prospective. POPULATION Between February 2019 and May 2021, 46 control (23 males, 49.3 ± 14.1 years), 45 suspected (20 males, 48.7 ± 12.5 years), and 46 definite (20 males, 53.7 ± 14.6 years) CP patients were enrolled at seven hospitals enrolled in the MINIMAP study. CP classification was based on imaging findings and clinical presentation. FIELD STRENGTH AND SEQUENCES 1.5 T. T1-weighted (T1W) spoiled gradient echo, T1 map with variable flip angle, dual-echo Dixon, secretin-enhanced MRCP before and after secretin infusion. ASSESSMENT Dual-echo fat fraction (FF), T1 relaxation time, extracellular volume (ECV), T1 signal intensity ratio of the pancreas to the spleen (T1 score), arterial-to-venous enhancement ratio (AVR), pancreatic tail diameter (PTD), pancreas volume, late gadolinium enhancement, pancreatic ductal elasticity (PDE), and duodenal filling grade of secretin-enhanced MRCP were measured. STATISTICAL TESTS Logistic regression analysis generated CP-MRI and secretin-enhanced CP-SMRI scores. Receiver operating characteristics analysis was used to differentiate definite CP from control. Interobserver agreement was assessed using Lin's concordance correlation coefficient. RESULTS Compared to control, definite CP cohort showed significantly higher dual-echo FF (7% vs. 11%), lower AVR (1.35 vs. 0.85), smaller PTD (2.5 cm vs. 1.95 cm), higher ECV (28% vs. 38%), and higher incidence of PDE loss (6.5% vs. 50%). With the cut-off of >2.5 CP-MRI score (dual-echo FF, AVR, and PTD) and CP-SMRI score (dual-echo FF, AVR, PTD, and PDE) had cross-validated area under the curves of 0.84 (sensitivity 87%, specificity 68%) and 0.86 (sensitivity 89%, specificity 67%), respectively. Interobserver agreement for both CP-MRI and CP-SMRI scores was 0.74. CONCLUSION The CP-MRI and CP-SMRI scores yielded acceptable performance and interobserver agreement for the diagnosis of CP. EVIDENCE LEVEL 1 TECHNICAL EFFICACY: Stage 2.
Collapse
Grants
- U01DK108323 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108288 NIDDK NIH HHS
- U01 DK108323 NIDDK NIH HHS
- U01DK108306 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108328 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108300 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01DK108327 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108327 NIDDK NIH HHS
- U01DK108288 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- DKP3041301 The Consortium for the Study of Chronic Pancreatitis, Diabetes, and Pancreatic Cancer
- U01 DK108300 NIDDK NIH HHS
- R01 DK116963 NIDDK NIH HHS
- U01 DK108306 NIDDK NIH HHS
- U01 DK108328 NIDDK NIH HHS
- R01DK116963 NIDDK NIH HHS
- R01DK116963 NIDDK NIH HHS
- National Institute of Diabetes and Digestive and Kidney Diseases
Collapse
Affiliation(s)
- Temel Tirkes
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Dhiraj Yadav
- Division of Gastroenterology, Hepatology & Nutrition, Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Darwin L. Conwell
- Department of Internal MedicineUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Xuandong Zhao
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Anil K. Dasyam
- Department of RadiologyUniversity of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Vivek Gowdra Halappa
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Aashish Patel
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | - Zarine K. Shah
- Department of RadiologyThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Jordan Swensson
- Department of Radiology and Imaging SciencesIndiana University School of MedicineIndianapolisIndianaUSA
| | | | | | - Ashley Wachsman
- Department of Imaging, Cedars‐Sinai Medical CenterUniversity of California in Los AngelesLos AngelesCaliforniaUSA
| | - Liang Li
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Kristofer Jennings
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Yunlong Yang
- Department of BiostatisticsThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Phil A. Hart
- Division of Gastroenterology, Hepatology & NutritionThe Ohio State University Wexner Medical CenterColumbusOhioUSA
| | - Stephen J. Pandol
- Division of Digestive and Liver DiseasesCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Walter G. Park
- Division of Gastroenterology and Hepatology, Department of MedicineStanford University Medical CenterStanfordCaliforniaUSA
| | | | | | - Paul R. Territo
- Division of Clinical PharmacologyStark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Scott A. Persohn
- Stark Neurosciences Research Institute, Indiana University School of MedicineIndianapolisIndianaUSA
| | - Dana K. Andersen
- Division of Digestive Diseases and NutritionNational Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of HealthBethesdaMarylandUSA
| | - Evan L. Fogel
- Lehman, Bucksot and Sherman Section of Pancreatobiliary EndoscopyIndiana University School of MedicineIndianapolisIndianaUSA
| | | |
Collapse
|
206
|
Tang L, Hill MC, He M, Chen J, Wang Z, Ellinor PT, Li M. A 3D Genome Atlas of Genetic Variants and Their Pathological Effects in Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408420. [PMID: 40134047 PMCID: PMC12097094 DOI: 10.1002/advs.202408420] [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] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 03/03/2025] [Indexed: 03/27/2025]
Abstract
The hierarchical organization of the eukaryotic genome is crucial for nuclear activities and cellular development. Genetic aberrations can disrupt this 3D genomic architecture, potentially driving oncogenesis. However, current research often lacks a comprehensive perspective, focusing on specific mutation types and singular 3D structural levels. Here, pathological changes from chromosomes to nucleotides are systematically cataloged, including 10 789 interchromosomal translocations (ICTs), 18 863 structural variants (SVs), and 162 769 single nucleotide polymorphisms (SNPs). The multilayered analysis reveals that fewer than 10% of ICTs disrupt territories via potent 3D interactions, and only a minimal fraction of SVs disrupt compartments or intersect topologically associated domain structures, yet these events significantly influence gene expression. Pathogenic SNPs typically show reduced interactions within the 3D genomic space. To investigate the effects of variants in the context of 3D organization, a two-phase scoring algorithm, 3DFunc, is developed to evaluate the pathogenicity of variant-gene pairs in cancer. Using 3DFunc, IGHV3-23's critical role in chronic lymphocytic leukemia is identified and it is found that three pathological SNPs (rs6605578, rs7814783, rs2738144) interact with DEFA3. Additionally, 3DGAtlas is introduced, which provides a highly accessible 3D genome atlas and a valuable resource for exploring the pathological effects of genetic mutations in cancer.
Collapse
Affiliation(s)
- Li Tang
- School of Computer Science and EngineeringCentral South UniversityChangsha410083China
| | - Matthew C. Hill
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA02129USA
- Cardiovascular Disease InitiativeThe Broad Institute of MIT and HarvardCambridgeMA02142USA
| | - Mingxing He
- School of Computer Science and EngineeringCentral South UniversityChangsha410083China
| | - Junhao Chen
- School of Computer Science and EngineeringCentral South UniversityChangsha410083China
| | - Zirui Wang
- School of Computer Science and EngineeringCentral South UniversityChangsha410083China
| | - Patrick T. Ellinor
- Cardiovascular Research CenterMassachusetts General HospitalBostonMA02129USA
- Cardiovascular Disease InitiativeThe Broad Institute of MIT and HarvardCambridgeMA02142USA
| | - Min Li
- School of Computer Science and EngineeringCentral South UniversityChangsha410083China
| |
Collapse
|
207
|
Yonghao O, Li Z, Xiao Y, Cai L, Liao Y, Lu D, Zhu H, Shen R, Wang X. The Survival Benefits of Chemotherapy for Undifferentiated Carcinoma With Osteoclast-Like Giant Cells of Pancreas: A Retrospective Analysis and Individual Participant Data Meta-Analysis. Cancer Med 2025; 14:e70894. [PMID: 40347088 PMCID: PMC12065096 DOI: 10.1002/cam4.70894] [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: 11/01/2024] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 05/12/2025] Open
Abstract
BACKGROUND AND AIM Undifferentiated carcinoma with osteoclast-like giant cells of pancreas (UCOGCP) is a rare type of pancreatic adenocarcinoma (PDAC). However, the efficacy of chemotherapy in UCOGCP has not been evaluated so far due to the scarcity of cases. This study aims to evaluate the efficacy of chemotherapy in UCOGCP combined with previous individual participant data (IPD) and SEER database data. METHODS Forty-nine patients with UCOGCP were enrolled from the Surveillance, Epidemiology, and End Results (SEER) database. Based on whether they had received chemotherapy or not, we divided UCOGCP patients into chemotherapy group (N = 32) and non-chemotherapy group (N = 17). The survival time of the chemotherapy group and non-chemotherapy group was assessed by Kaplan-Meier analysis and Cox analysis. IPD data for UCOGCP were collected in PubMed, Embase, Cochrane, and ScienceDirect. The results based on the SEER database were verified by IPD meta-analysis. RESULTS The Kaplan-Meier analysis indicated that patients who received chemotherapy experienced a longer survival time compared to those who did not (OS: p = 0.00061, CSS: p = 0.00047). Univariate (OS: HR: 0.31 [0.15, 0.63], p = 0.001; CSS: HR: 0.28 [0.13, 0.60], p = 0.001) and multivariate (OS: HR: 0.33 [0.14, 0.78], p = 0.012; CSS: HR: 0.30 [0.12, 0.73], p = 0.008) Cox regression showed that chemotherapy was the independent prognostic protective factor for UCOGCP. IPD meta-analysis showed that chemotherapy can significantly improve the prognosis of patients who received primary tumor resection (PTR, p = 0.0084). CONCLUSION In contrast to not receiving chemotherapy, chemotherapy is effective in prolonging survival in UCOGCP patients with or without PTR. This provides a foundation for the use of UCOGCP chemotherapy.
Collapse
Affiliation(s)
- Ouyang Yonghao
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Zhi Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yi Xiao
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Lina Cai
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Yannian Liao
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Denghuan Lu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Hong Zhu
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Rongxi Shen
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Xinbo Wang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
- Department of General Surgery, Jinling Hospital, the First School of Clinical MedicineSouthern Medical UniversityNanjingChina
| |
Collapse
|
208
|
Abeysundara N, Rasnitsyn A, Fong V, Bahcheli A, Van Ommeren R, Juraschka K, Vladoiu M, Ong W, Livingston B, de Antonellis P, Ly M, Holgado BL, Sirbu O, Bahrampour S, Min HK, Fan J, Nor C, Visvanathan A, Zhang J, Wang H, Qin L, Huang N, Pallotta J, Douglas T, Mak E, Su H, Ng K, Zhang KY, Daniels C, Lucas CHG, Eberhart CG, Liu H, Jiang T, Notta F, Ramaswamy V, Reimand J, Gallo M, Rich JN, Wu X, Huang X, Taylor MD. Metastatic medulloblastoma remodels the local leptomeningeal microenvironment to promote further metastatic colonization and growth. Nat Cell Biol 2025; 27:863-874. [PMID: 40263572 PMCID: PMC12081294 DOI: 10.1038/s41556-025-01660-7] [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: 06/04/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Leptomeningeal metastases are the major source of morbidity and mortality for patients with medulloblastoma. The biology of the leptomeningeal metastases and the local tumour microenvironment are poorly characterized. Here we show that metastasis-associated meningeal fibroblasts (MB-MAFs) are transcriptionally distinct and signal extensively to tumour cells and the tumour microenvironment. Metastatic cells secrete platelet-derived growth factor (PDGF) ligands into the local microenvironment to chemotactically recruit meningeal fibroblasts. Meningeal fibroblasts are reprogrammed to become MB-MAFs, expressing distinct transcriptomes and secretomes, including bone morphogenetic proteins. Active bone morphogenetic protein signalling and co-implantation of tumour cells with MB-MAFs enhances the colonization of the leptomeninges by medulloblastoma cells and promotes the growth of established metastases. Furthermore, treatment of patient-derived xenograft mice with a PDGF-receptor-α neutralizing antibody enhances overall survival in vivo. Collectively, our results define a targetable intercellular communication cascade in the metastatic niche to treat leptomeningeal disease.
Collapse
Affiliation(s)
- Namal Abeysundara
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Alexandra Rasnitsyn
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Vernon Fong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alexander Bahcheli
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Randy Van Ommeren
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kyle Juraschka
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Maria Vladoiu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Winnie Ong
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Bryn Livingston
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Pasqualino de Antonellis
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Michelle Ly
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Borja López Holgado
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Olga Sirbu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Shahrzad Bahrampour
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Hyun-Kee Min
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Jerry Fan
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Carolina Nor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Abhirami Visvanathan
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jiao Zhang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Texas Children's Cancer and Hematology Center, Houston, TX, USA
- Department of Pediatrics - Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Hao Wang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lei Qin
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ning Huang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jonelle Pallotta
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tajana Douglas
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Esta Mak
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Haipeng Su
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Karen Ng
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Kevin Yang Zhang
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Craig Daniels
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Texas Children's Cancer and Hematology Center, Houston, TX, USA
- Department of Pediatrics - Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | | | - Charles G Eberhart
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hailong Liu
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Faiyaz Notta
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Vijay Ramaswamy
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jüri Reimand
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marco Gallo
- Texas Children's Cancer and Hematology Center, Houston, TX, USA
- Department of Pediatrics - Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Jeremy N Rich
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Xiaochong Wu
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Texas Children's Cancer and Hematology Center, Houston, TX, USA
- Department of Pediatrics - Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Xi Huang
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Michael D Taylor
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
- Texas Children's Cancer and Hematology Center, Houston, TX, USA.
- Department of Pediatrics - Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada.
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, USA.
- Department of Neurosurgery, Texas Children's Hospital, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
209
|
Mcneil TR, Sikder S, Dalal Y. Cancer cells' chamber of secrets: the link between micronuclei, chromothripsis and malignancy. Open Biol 2025; 15:240388. [PMID: 40359993 DOI: 10.1098/rsob.240388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/19/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Micronuclei exhibit defective proteomes rendering their chromatin vulnerable to fragmentation. This fragmentation process, known as chromothripsis, promotes tumorigenesis by catalysing the activation of oncogenes and the silencing of tumor suppressors. With this role in mind, micronuclei serve as promising targets for therapeutic intervention. This review will explore recent discoveries regarding how micronuclei form, their function in catalysing chromothripsis and how chromothripsis provides a selective advantage for cancer cells.
Collapse
Affiliation(s)
| | - Sweta Sikder
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
210
|
Yang S, He Y, Ma Y, Wang R, Wu Y, Wu W. Associations between the oral microbiome, number of teeth and frailty among American adults: A cross-sectional study from NHANES 2009-2012. Exp Gerontol 2025; 203:112727. [PMID: 40054511 DOI: 10.1016/j.exger.2025.112727] [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: 12/23/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The intricate interrelationship between oral health, the number of teeth, oral microbiota, and frailty remains largely unexplored in clinical research. This study aimed to investigate the interrelationship between oral microbiome, the number of teeth, and frailty. METHODS Data from 4518 participants in NHANES 2009-2012 were analyzed. Frailty was measured using the 48-item Frailty Index (FI). Multivariable logistic regression and restricted cubic spline (RCS) evaluated associations between alpha diversity and frailty. Mediation analysis was used to assess the role of number of teeth. The associations between oral microbiome diveristy and mortality were analyzed by Cox regression. Beta diversity was examined with PCoA and PERMANOVA. RESULTS The prevalence of frailty was 39.73 %. Univariate analysis showed that alpha diversity indices except for the Simpson index were significantly lower in frailty, and after adjusted for confounders, observed ASVs (adjusted OR: 0.80 [0.73, 0.87], p < 0.001), Faith's PD (adjusted OR: 0.81 [0.74, 0.88], p < 0.001) and Shannon-Weiner index (adjusted OR: 0.88 [0.81, 0.95], p = 0.002) were remained significantly associated with frailty. The reduced number of teeth partially mediated the relationship (for Faith's PD: βindirect = -0.001 [-0.003, 0.000], p = 0.036, proportion: 8.33 % [0.00 %, 37.50 %]; for Shannon-Weiner index, βindirect = -0.007 [-0.013, -0.002], p = 0.007, Proportion = 17.07 % [3.39 %, 65.00 %]). Univariable Cox proportional hazard regression showed that all alpha diversity indices were significantly associated with all-cause mortality in frail population, and in multivariable analysis, Shannon-Weiner index (HR: 0.72 [0.55, 0.94], p = 0.017) and Simpson index (HR: 0.71 [0.60, 0.83], p < 0.001) remained statistically significant. PCoA showed that beta diversity was also significantly associated with frailty. CONCLUSION Lower oral microbiome diversity is associated with higher frailty and mortality. The number of teeth partially mediates this link, emphasizing the importance of oral health in mitigating frailty and promoting healthy aging.
Collapse
Affiliation(s)
- Sixiang Yang
- School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China
| | - Yanyun He
- School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China
| | - Yuping Ma
- School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China
| | - Ruoli Wang
- School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China
| | - Yeke Wu
- Department of Stomatology, Hospital of Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China
| | - Wenbin Wu
- School of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Shi'erqiao Road, Chengdu 610075, Sichuan, China.
| |
Collapse
|
211
|
Elsayed B, Elshoeibi AM, Elhadary M, Al-Jubouri AM, Al-Qahtani N, Vranic S, Al-Saady R. Molecular and immunohistochemical markers in appendiceal mucinous neoplasms: A systematic review and comparative analysis with ovarian mucinous neoplasms and colorectal adenocarcinoma. Histol Histopathol 2025; 40:621-633. [PMID: 39743929 DOI: 10.14670/hh-18-830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
INTRODUCTION Appendiceal mucinous neoplasms (AMNs) represent a rare and diagnostically challenging group of tumors. This systematic review aims to summarize the reported molecular and immunohistochemical markers (IHC) associated with AMNs and compare them with ovarian mucinous neoplasms (OMNs) and colorectal adenocarcinoma (CRC). METHODS A comprehensive search was performed in PubMed/MEDLINE/PMC, Scopus, Embase, and Web of Science databases to identify studies looking at IHC and molecular markers in AMNs. Chi-squared and Fisher's exact tests were utilized to compare the marker expression across different tumor types. RESULTS We identified 27 articles reporting several potential biomarkers for distinguishing between different subtypes of AMNs. Mutations in KRAS, GNAS, and RNF43 emerged as notable biomarkers, with KRAS mutations being the most prevalent across all subtypes. Additionally, p53 IHC overexpression was associated with higher tumor grades. When comparing AMNs with OMNs, we observed a higher prevalence of CK20, CDX2, SATB2, and MUC2 IHC expression, as well as KRAS and GNAS mutations, in AMNs. Conversely, CK7 and PAX8 IHC expression were more prevalent in OMNs. Comparing AMNs with CRCs, we found a higher prevalence of TOPO1 and PTEN IHC expression, as well as KRAS and GNAS mutations, in AMNs. Conversely, nuclear β-catenin IHC expression, as well as TP53, APC, and PIK3CA mutations, were more prevalent in CRCs. CONCLUSION This systematic review identified possible markers for distinguishing AMNs and differentiating between AMNs, OMNs, or CRCs.
Collapse
Affiliation(s)
- Basel Elsayed
- College of Medicine, QU Health, Qatar University, Doha, Qatar.
| | | | | | | | - Noof Al-Qahtani
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Semir Vranic
- Department of Pathology, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rafif Al-Saady
- Department of Pathology, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
212
|
Christe A, Primetis E, Cereghetti GM, Drakopoulos D, Dhayat NA, Bonny O, Ritter A, Mohebbi N, Faller N, Pellegrini L, Bedino G, Venzin RM, Grosse P, Hüsler C, Koneth I, Bucher C, Del Giorno R, Gabutti L, Mayr M, Odermatt U, Buchkremer F, Ernandez T, Stoermann-Chopard C, Teta D, Tamò L, Trelle S, Roth B, Bargagli M, Fuster DG. Hydrochlorothiazide and Bone Mineral Density in Patients with Kidney Stones: Post Hoc Analysis of the NOSTONE Trial. Clin J Am Soc Nephrol 2025; 20:706-718. [PMID: 40063114 PMCID: PMC12097187 DOI: 10.2215/cjn.0000000659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
Key Points Loss of bone mineral density at 3 years was similar in patients with calcium kidney stones randomized to hydrochlorothiazide or placebo. There was no association between hydrochlorothiazide dose and change in bone mineral density at 3 years. Results were consistent across sensitivity and per-protocol analyses. Background Low bone mass and fractures are common among kidney stone formers, yet it remains unclear whether thiazides can help preserve bone mass. We aimed to evaluate the effectiveness of a range of hydrochlorothiazide (HCTZ) doses compared with a placebo on bone mineral density (BMD) over a 3-year period. Methods This post hoc analysis was conducted on data from the NOSTONE trial, a multicenter, randomized, controlled study. A total of 416 adults with recurrent calcium stones participated in the study, receiving either placebo or HCTZ at doses of 12.5, 25, or 50 mg daily. BMD was measured using computed tomography at the T12–L3 vertebrae at both baseline and the end of the study. Results Over a median follow-up period of 2.92 years, the mean BMD decreased by 6.4±15.7 Hounsfield units (HU) in the placebo group, 5.1±15.1 HU in the 12.5 mg HCTZ group (β coefficient versus placebo, 0.37 HU; 95% confidence interval [CI], −1.74 to 2.47; P = 0.73), 4.1±16.3 HU in the 25 mg HCTZ group (β , 0.93 HU; 95% CI, −1.34 to 3.19; P = 0.42), and 4.8±15.9 HU in the 50 mg HCTZ group (β , 0.70 HU; 95% CI, −1.45 to 2.85; P = 0.52). No association was observed between HCTZ dose and BMD at the end of the study (P = 0.43). The results were confirmed in sensitivity analyses for eGFR, urine calcium, net gastrointestinal alkali absorption, and body mass index; in subgroup; and in per-protocol analyses. Conclusions In patients with recurrent calcium kidney stones, loss of BMD was similar in patients receiving HCTZ at a dose of 12.5, 25, or 50 mg or placebo once daily. Clinical Trial registry name and registration number: NCT03057431 .
Collapse
Affiliation(s)
- Andreas Christe
- Department of Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Elias Primetis
- Department of Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Grazia M. Cereghetti
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Dionysios Drakopoulos
- Department of Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Nasser A. Dhayat
- Nephrology and Dialysis Care Center, B. Braun Medical Care AG, Hochfelden, Zürich, Switzerland
| | - Olivier Bonny
- Service of Nephrology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Service of Nephrology, Fribourg State Hospital and University of Fribourg, Fribourg, Switzerland
| | - Alexander Ritter
- Department of Nephrology, University Hospital Zürich, Zürich, Switzerland
| | - Nilufar Mohebbi
- Department of Nephrology, University Hospital Zürich, Zürich, Switzerland
| | - Nicolas Faller
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lisa Pellegrini
- Department of Nephrology, Regional Hospital Lugano, Lugano, Switzerland
| | - Giulia Bedino
- Department of Nephrology, Regional Hospital Lugano, Lugano, Switzerland
| | - Reto M. Venzin
- Department of Nephrology, Cantonal Hospital Graubünden, Chur, Switzerland
| | - Philipp Grosse
- Department of Nephrology, Cantonal Hospital Graubünden, Chur, Switzerland
| | - Carina Hüsler
- Department of Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Irene Koneth
- Department of Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Christian Bucher
- Department of Nephrology and Transplantation Medicine, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Rosaria Del Giorno
- Department of Internal Medicine, Regional Hospital Bellinzona, Bellinzona, Switzerland
| | - Luca Gabutti
- Department of Internal Medicine, Regional Hospital Bellinzona, Bellinzona, Switzerland
| | - Michael Mayr
- Medical Outpatient Department, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Urs Odermatt
- Department of Nephrology, Cantonal Hospital Luzern, Luzern, Switzerland
| | - Florian Buchkremer
- Division of Nephrology, Dialysis and Transplantation, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Thomas Ernandez
- Department of Nephrology, HUG, University Hospital Geneva, University of Geneva, Geneva, Switzerland
| | | | - Daniel Teta
- Service de Nephrology, Centre Hospitalier du Valais Romand, CHVR, Sion, Switzerland
| | - Luca Tamò
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sven Trelle
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Beat Roth
- Department of Urology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Matteo Bargagli
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel G. Fuster
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
213
|
Wyzlic P, Damanakis A, Quaas A, Bruns CJ, Schmidt T. [Relevance of frozen section diagnostics in pancreatic surgery]. CHIRURGIE (HEIDELBERG, GERMANY) 2025; 96:371-377. [PMID: 40063096 DOI: 10.1007/s00104-025-02265-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 04/23/2025]
Abstract
Frozen sections are performed in pancreatic surgery for three reasons: histopathological confirmation of previously unclear space-occupying lesions, determination of the extent of surgical resection in an operative exploration and for possible follow-up resections after previously carried out surgical resections. Overall, in the literature there are heterogeneous data with respect to an improvement in the prognosis of a secondary R0 resection by a repeat resection in comparison to a R1 resection. Nowadays, extended pancreatic resections including vascular resections are technically feasible and safe. Nevertheless, with respect to the precise radicality in the surgical procedure, all patient characteristics should be taken into consideration in addition to the histopathological diagnosis of the frozen sections.
Collapse
Affiliation(s)
- Patricia Wyzlic
- Klinik und Poliklinik für Allgemein‑, Viszeral‑, Thorax- und Transplantationschirurgie, Uniklinikum Köln, Kerpener Straße 62, 50937, Köln, Deutschland
| | - Alexander Damanakis
- Klinik und Poliklinik für Allgemein‑, Viszeral‑, Thorax- und Transplantationschirurgie, Uniklinikum Köln, Kerpener Straße 62, 50937, Köln, Deutschland
| | - Alexander Quaas
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Uniklinikum Köln, Köln, Deutschland
| | - Christiane J Bruns
- Klinik und Poliklinik für Allgemein‑, Viszeral‑, Thorax- und Transplantationschirurgie, Uniklinikum Köln, Kerpener Straße 62, 50937, Köln, Deutschland
| | - Thomas Schmidt
- Klinik und Poliklinik für Allgemein‑, Viszeral‑, Thorax- und Transplantationschirurgie, Uniklinikum Köln, Kerpener Straße 62, 50937, Köln, Deutschland.
| |
Collapse
|
214
|
Edelbach B, Glaser D, Almekkawi AK, Caruso JP, Sbaiti G, Aoun SG, Bagley CA. Optimal Duration of Antibiotic Therapy for Primary Osteomyelitis Discitis: A Systematic Review and Network Meta-Analysis. Spine (Phila Pa 1976) 2025; 50:636-644. [PMID: 39722225 DOI: 10.1097/brs.0000000000005244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
STUDY DESIGN Systematic review and network meta-analysis. OBJECTIVE This study aimed to systematically review the literature on the management of primary osteomyelitis discitis and perform a network meta-analysis comparing the efficacy of different antibiotic treatment durations. BACKGROUND Primary osteomyelitis discitis is a challenging condition with varying management strategies. MATERIALS AND METHODS A comprehensive literature search was conducted. Studies reporting outcomes for the treatment of primary osteomyelitis discitis were included. A random-effect network meta-analysis was performed comparing antibiotic treatment durations of <4 weeks, 4 to 8 weeks, 8 to 12 weeks, and 12 to 16 weeks. The surface under the cumulative ranking curve (SUCRA) was used to rank treatment effectiveness. RESULTS Sixty-three articles with 4233 patients were included. Staphylococcus aureus was the most common causative agent (57.6%). The 4 to 8-week antibiotic duration ranked highest across fixed-effect and random-effect models (SUCRA: 0.8207 and 0.8343). The 12 to 16-week duration ranked highest in the fixed-effect model (SUCRA: 0.8460) but dropped substantially in the random-effect model (SUCRA: 0.3067). The <4-week duration showed mixed results. The 8 to 12-week duration consistently ranked lowest. No statistically significant differences were found between durations for symptomatic relief. CONCLUSION Antibiotic therapy for 4 to 8 weeks may provide the optimal balance of efficacy and treatment duration for most patients with primary osteomyelitis discitis. However, treatment should be individualized based on clinical response. Further prospective studies are needed to clarify optimal management strategies for this complex condition.
Collapse
Affiliation(s)
| | - Dylan Glaser
- Department of Neurosurgery, School of Medicine, University of Missouri-Kansas City
| | - Ahmad K Almekkawi
- Department of Neurosurgery, Saint Luke's Marion Bloch Neuroscience Institute, Kansas City, MO
| | - James P Caruso
- Department of Neurosurgery, The University of Texas Southwestern, Dallas, TX
| | - Ghewa Sbaiti
- Department of Pharmacy, The University of Texas Southwestern, Dallas, TX
| | - Salah G Aoun
- Department of Neurosurgery, The University of Texas Southwestern, Dallas, TX
| | - Carlos A Bagley
- Department of Neurosurgery, School of Medicine, University of Missouri-Kansas City
- Department of Neurosurgery, Saint Luke's Marion Bloch Neuroscience Institute, Kansas City, MO
| |
Collapse
|
215
|
Powrózek T, Otieno MO, Maffeo D, Frullanti E, Martinez-Useros J. Blood circulating miRNAs as pancreatic cancer biomarkers: An evidence from pooled analysis and bioinformatics study. Int J Biol Macromol 2025; 308:142469. [PMID: 40180095 DOI: 10.1016/j.ijbiomac.2025.142469] [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: 08/08/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer (PC) is one of the deadliest cancers, characterized by a poor prognosis. Currently, there are no screening programs for the early detection of PC, and existing diagnostic methods are primarily limited to high-risk individuals. Biomarkers such as CA19-9 have not significantly improved early diagnosis, making the identification of new potential biomarkers crucial for routine clinical practice. Among the candidate biomarkers, miRNAs have been most extensively studied due to their role in regulating gene expression (either as oncomiRs or tumor suppressor miRNAs) and their potential for minimally invasive analysis through liquid biopsy techniques. This review aims to summarize the current literature on blood-circulating miRNAs and their diagnostic value in PC detection, considering the context of CA19-9 and benign pancreatic diseases. The data from the collected studies were curated through both statistical and bioinformatics analyses to identify the most promising miRNAs with optimal diagnostic accuracy for PC detection and to assess their role in the molecular processes leading to tumor development.
Collapse
Affiliation(s)
- Tomasz Powrózek
- Department of Human Physiology, Medical University of Lublin, Lublin, Poland.
| | - Michael Ochieng' Otieno
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain
| | - Debora Maffeo
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elisa Frullanti
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Cancer Genomics and Systems Biology Lab, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Javier Martinez-Useros
- Translational Oncology Division, Oncohealth Institute, Fundacion Jiménez Díaz University Hospital, Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Madrid, Spain
| |
Collapse
|
216
|
Ye L, Fei Q, Li T, Lin C, Ou H, Zhang Y, Zhou Y, Zhang J, Jin K, Yu X, Wu W. Genomic mutation and Clinicopathological features of KRAS wild-type tumors in Chinese cohort with pancreatic adenocarcinoma. Genomics 2025; 117:111048. [PMID: 40274200 DOI: 10.1016/j.ygeno.2025.111048] [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: 12/19/2024] [Revised: 03/31/2025] [Accepted: 04/17/2025] [Indexed: 04/26/2025]
Abstract
This study aims to elucidate the genomic and clinicopathological characteristics of pancreatic ductal adenocarcinoma (PDAC) in Chinese patients with KRAS wild-type (WT) tumors. Analysis of 869 PDAC patients revealed that 164 tumors (19 %) were KRAS WT, with a predominance of TP53 mutations (32 %), followed by AHNAK (12 %), CTNNB1 (6 %), and BRAF (5 %). These tumors exhibited a higher prevalence of DNA damage response gene mutations, lower levels of tumor antigens CA-199, CA-125, and CEA, and with a similar trend in mutant-allele tumor heterogeneity and tumor mutational burden. Conversely, microsatellite instability was markedly elevated in these cases. Survival outcomes were superior for KRAS WT tumors, with a median overall survival of 36.5 months compared to 23.0 months for KRAS mutated tumors (P < 0.0001). These findings suggest that KRAS WT PDAC in Chinese patients presents a distinct genetic profile necessitating the development of specific therapeutic strategies.
Collapse
Affiliation(s)
- Longyun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Qinglin Fei
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Tianjiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Chang Lin
- Zhejiang Chinese Medical University, Jiaxing 314001, Zhejiang Province, China
| | - Huiyi Ou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yiting Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Yi Zhou
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China
| | - Jie Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Hospital of Jiaxing, 1882 South Central Road, South Lake., Jiaxing 314001, Zhejiang Province, China.
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| | - Weiding Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Shanghai Pancreatic Cancer Institute, Shanghai 200032, China; Pancreatic Cancer Institute, Fudan University, Shanghai 200032, China.
| |
Collapse
|
217
|
Boreskie PE, Boreskie KF. Frailty-aware Care in the Emergency Department. Emerg Med Clin North Am 2025; 43:199-210. [PMID: 40210341 DOI: 10.1016/j.emc.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Older adults living with frailty are a growing population that will increasingly present to the emergency department (ED). This is a population that is at increased risk of adverse health outcomes and most EDs are not designed with their needs in mind. Instead of characterizing patients based on chronologic age or existing triage tools, frailty assessment offers an accurate, feasible, and patient-centered approach to improving care, and should be performed in the ED.
Collapse
Affiliation(s)
- Patrick E Boreskie
- Department of Emergency Medicine, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Emergency Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Kevin F Boreskie
- Department of Emergency Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Applied Health Sciences, Faculty of Graduate Studies, University of Manitoba, Winnipeg, Manitoba, Canada; University of Manitoba (Bannatyne campus), S203 Medical Services Building, 750 Bannatyne Avenue, Winnipeg, MB R3E 0W2, Canada
| |
Collapse
|
218
|
Laosuwan K, Vichiansiri R, Somboonporn C, Saengsuwan J. Accuracy of four sarcopenia screening methods in patients with chronic stroke in Thailand: a cross-sectional study. Top Stroke Rehabil 2025; 32:371-381. [PMID: 39344776 DOI: 10.1080/10749357.2024.2409000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
AIMS The aim of this study was to evaluate the accuracy of screening tools for sarcopenia and to determine whether the same or different cutoff points should be applied in patients with chronic stroke. MATERIALS AND METHODS Sixty-eight participants with residual hemiparetic deficit for over 6 months were enrolled. We evaluated the accuracy of calf circumference, SARC-F questionnaire, SARC-CalF, and Ishii's score chart using the Asia Working Group for Sarcopenia (AWGS) 2019 revised criteria as the gold standard. RESULTS Sarcopenia was identified in 22 participants (32.4%) based on the AWGS criteria. Overall, SARC-F showed the lowest diagnostic accuracy. The Area Under the receiver operating characteristic Curves (AUC) of calf circumference, SARC-F, SARC-CalF, and Ishii's score chart were 0.77 (95% confidence interval [CI], 0.66-0.88), 0.58 (95% CI, 0.42-0.74), 0.75 (95% CI, 0.62-0.87), and 0.78 (95% CI, 0.65-0.90), respectively. The mean AUC of SARC-F was inferior to SARC-CalF (0.58 vs. 0.75, p = 0.035). CONCLUSIONS The accuracy and diagnostic properties of calf circumference, SARC-CalF, and Ishii's score chart were comparable (mean AUC of 0.77, 0.75, and 0.78, respectively). SARC-F showed the lowest accuracy (mean AUC = 0.58). The recommended screening tools are calf circumference, SARC-CalF, and Ishii's score chart. It is not recommended to rely solely on SARC-F for screening sarcopenia after stroke. We proposed potential new cutoff points for each screening instrument, including SARC-F, SARC-CalF, calf circumference in women, and Ishii's score chart for both men and women.
Collapse
Affiliation(s)
- Kannanat Laosuwan
- Department of Rehabilitation Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ratana Vichiansiri
- Department of Rehabilitation Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Charoonsak Somboonporn
- Nuclear Medicine Division, Department of Radiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jittima Saengsuwan
- Department of Rehabilitation Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- North-Eastern Stroke Research Group, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
219
|
Eikenboom EL, Nasar N, Seier K, Gönen M, Spaander MCW, O'Reilly EM, Jarnagin WR, Drebin J, D'Angelica MI, Kingham TP, Balachandran VP, Soares KC, Wagner A, Wei AC. Survival of Patients with Resected Microsatellite Instability-High, Mismatch Repair Deficient, and Lynch Syndrome-Associated Pancreatic Ductal Adenocarcinomas. Ann Surg Oncol 2025; 32:3568-3577. [PMID: 39656390 DOI: 10.1245/s10434-024-16621-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: 06/07/2024] [Accepted: 11/19/2024] [Indexed: 04/24/2025]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) remains a challenging disease due to its aggressiveness, late-stage diagnosis, and limited treatment options. Microsatellite instability-high (MSI-H) cancers are susceptible to immune checkpoint inhibitors. Survival outcomes for patients with MSI-H PDAC are unknown as the disease is rare. METHODS This study included patients with PDACs surgically resected from 1990 to 2023, and those with germline or sporadic pathogenic variants in DNA mismatch repair genes were identified. The study matched MSI-H, mismatch repair-deficient (MMRd), and Lynch syndrome (LS)-associated PDAC cases (on age, gender, and year of surgery) with microsatellite-stable (MSS), mismatch repair-proficient, or non-LS-associated PDAC cases in a 1:2 ratio. A generalized estimating equation Cox model with a robust sandwich estimator was used to compare overall survival (OS) in the matched cohorts. RESULTS Of 936 cases, 18 were included. Eight cases were MSI-H/MMRd, two were MSI/IHC-indeterminate, seven were MSS, and one was not tested for MSI. Nine patients had LS (MLH1 [n = 1], MSH2 [n = 4], MSH6 [n = 1], PMS2 [n = 3]), and nine patients had sporadic pathogenic variants in DNA MMR genes (MLH1 [n = 4], MSH6 [n = 5]). After matching to 36 control patients, the MSI-H/MMRd/LS PDACs had a significantly better OS (hazard ratio [HR], 0.36 [95% confidence interval [CI], 0.18-0.73; p = 0.005]; 5-year OS: MSI-H 77% [95% CI 58-100%] vs. MSS 27% [95% CI 15-51%]). CONCLUSION Before routine use of immune checkpoint inhibitors, the patients with MSI-H, MMRd, and LS-associated PDACs displayed significantly better survival than the patients with MSS, MMR-proficient, non-LS-associated PDACs. It is expected that survival for this cohort will further improve with increased availability of immunotherapy.
Collapse
Affiliation(s)
- Ellis L Eikenboom
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Naaz Nasar
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth Seier
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey Drebin
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kevin C Soares
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anja Wagner
- Department of Clinical Genetics, Erasmus Medical Center Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Alice C Wei
- Hepato-Pancreato-Biliary Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
220
|
Chalepaki AM, Gkoris M, Chondrou I, Kourti M, Georgakopoulos-Soares I, Zaravinos A. A multi-omics analysis of effector and resting treg cells in pan-cancer. Comput Biol Med 2025; 189:110021. [PMID: 40088713 DOI: 10.1016/j.compbiomed.2025.110021] [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: 10/05/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025]
Abstract
Regulatory T cells (Tregs) are critical for maintaining the stability of the immune system and facilitating tumor escape through various mechanisms. Resting T cells are involved in cell-mediated immunity and remain in a resting state until stimulated, while effector T cells promote immune responses. Here, we investigated the roles of two gene signatures, one for resting Tregs (FOXP3 and IL2RA) and another for effector Tregs (FOXP3, CTLA-4, CCR8 and TNFRSF9) in pan-cancer. Using data from The Cancer Genome Atlas (TCGA), The Cancer Proteome Atlas (TCPA) and Gene Expression Omnibus (GEO), we focused on the expression profile of the two signatures, the existence of single nucleotide variants (SNVs) and copy number variants (CNVs), methylation, infiltration of immune cells in the tumor and sensitivity to different drugs. Our analysis revealed that both signatures are differentially expressed across different cancer types, and correlate with patient survival. Furthermore, both types of Tregs influence important pathways in cancer development and progression, like apoptosis, epithelial-to-mesenchymal transition (EMT) and the DNA damage pathway. Moreover, a positive correlation was highlighted between the expression of gene markers in both resting and effector Tregs and immune cell infiltration in adrenocortical carcinoma, while mutations in both signatures correlated with enrichment of specific immune cells, mainly in skin melanoma and endometrial cancer. In addition, we reveal the existence of widespread CNVs and hypomethylation affecting both Treg signatures in most cancer types. Last, we identified a few correlations between the expression of CCR8 and TNFRSF9 and sensitivity to several drugs, including COL-3, Chlorambucil and GSK1070916, in pan-cancer. Overall, these findings highlight new evidence that both Treg signatures are crucial regulators of cancer progression, providing potential clinical outcomes for cancer therapy.
Collapse
Affiliation(s)
- Anna-Maria Chalepaki
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Marios Gkoris
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| | - Irene Chondrou
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Malamati Kourti
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus.
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA.
| | - Apostolos Zaravinos
- Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia, Cyprus; Cancer Genetics, Genomics and Systems Biology Laboratory, Basic and Translational Cancer Research Center (BTCRC), Nicosia, Cyprus.
| |
Collapse
|
221
|
Iyer MK, Fletcher A, Okoye JO, Shi C, Chen F, Kanu E, Eckhoff AM, Bao M, di Magliano MP, Frankel TL, Chinnaiyan AM, Nussbaum DP, Allen PJ. Spatial Transcriptomics of Intraductal Papillary Mucinous Neoplasms Reveals Divergent Indolent and Malignant States. Clin Cancer Res 2025; 31:1796-1808. [PMID: 39969959 PMCID: PMC12045729 DOI: 10.1158/1078-0432.ccr-24-1529] [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: 05/23/2024] [Revised: 09/20/2024] [Accepted: 02/17/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Intraductal papillary mucinous neoplasms (IPMN) occur in 5% to 10% of the population, but only a small minority progress to pancreatic ductal adenocarcinoma (PDAC). The lack of accurate predictors of high-risk disease leads to both unnecessary operations for indolent neoplasms and missed diagnoses of PDAC. Digital spatial RNA profiling (DSP-RNA) provides an opportunity to define and associate transcriptomic states with cancer risk. EXPERIMENTAL DESIGN We performed whole-transcriptome DSP-RNA profiling on 10 IPMN specimens encompassing the spectrum of dysplastic changes from normal duct to cancer. Epithelial regions within each tissue were annotated as normal duct, low-grade dysplasia, high-grade dysplasia, or invasive carcinoma. The resulting digital gene expression data were analyzed with R/Bioconductor. RESULTS Our analysis uncovered three distinct epithelial transcriptomic states-"normal-like" (cNL), "low risk" (cLR), and "high risk" (cHR)-which were significantly associated with pathologic grade. Furthermore, the three states were significantly correlated with the exocrine, classical, and basal-like molecular subtypes described in PDAC. Specifically, exocrine function diminished in cHR, classical activation distinguished neoplasia (cLR and cHR) from cNL, and basal-like genes were specifically upregulated in cHR. Intriguingly, markers of cHR were detected in normal duct and low-grade dysplasia regions from specimens with PDAC but not from specimens containing only low-grade IPMN. CONCLUSIONS DSP-RNA of IPMN revealed low-risk (indolent) and high-risk (malignant) expression programs that correlated with the activity of exocrine and basal-like PDAC signatures, respectively, and distinguished pathologically low-grade specimens from malignant specimens. These findings contextualize IPMN pathogenesis and have the potential to improve risk stratification.
Collapse
Affiliation(s)
- Matthew K. Iyer
- Department of Surgery, Duke University; Durham, North Carolina
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Ashley Fletcher
- Department of Surgery, Duke University; Durham, North Carolina
| | - Jude Ogechukwu Okoye
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Histopathology, Nnamdi Azikiwe University, Nnewi, Nigeria
| | - Chanjuan Shi
- Department of Pathology, Duke University; Durham, North Carolina
| | - Fengming Chen
- Department of Pathology, Duke University; Durham, North Carolina
| | - Elishama Kanu
- Department of Surgery, Duke University; Durham, North Carolina
| | | | - Matthew Bao
- Department of Surgery, Duke University; Durham, North Carolina
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | | | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, Duke University; Durham, North Carolina
- Department of Urology, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | | | - Peter J. Allen
- Department of Surgery, Duke University; Durham, North Carolina
| |
Collapse
|
222
|
Nunna RS, Gruber MD, Karuparti S, Taylor Z, Genovese S, Jumah F, Godolias P, Tataryn Z, Hollern D, Oskouian R, Chapman JR. Cost-Effectiveness Analyses of Bone Morphogenetic Protein 2 (rhBMP-2) in Spinal Fusion: A Systematic Review. Global Spine J 2025; 15:2514-2535. [PMID: 39707701 PMCID: PMC11662347 DOI: 10.1177/21925682241310864] [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: 05/28/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024] Open
Abstract
Study DesignSystematic Review Study.ObjectivesThe objective of this study is to assess the cost-effectiveness and cost-utility of rhBMP-2 in spine surgeries.MethodsPubMed, MEDLINE, OVID and CENTRAL (Cochrane Central Register of Controlled Trials) databases were systematically searched for studies reporting the use of rhBMP-2 for spinal procedures in comparison to autograft and/or allograft and alternative graft materials under consideration of cost-effectiveness, cost-benefit, cost minimization, and/or cost-utility analysis. Relevant data points were extracted and compiled. The standardization of the selected studies was graded using the CHEERS criteria.ResultsOf the 88 eligible publications, 17 met the inclusion criteria. Overall, 8 studies showed a favorable cost-analysis profile. Three studies performed cost-consequence analyses, none of which reported positive findings. Among 7 cost-utility analyses and 7 cost-effectiveness analyses, 3 and 5, respectively, reported a likely benefit to the usage of rhBMP-2. The CHEERS scores for the included studies ranged from 15 to 23 out of 24 points, with a median score of 19.ConclusionsThere is no consensus regarding the cost-effectiveness or cost-utility of rhBMP-2 in spinal fusion. rhBMP-2 may function as a cost-effective adjunct in spinal fusion surgeries in select cases.
Collapse
Affiliation(s)
- Ravi S. Nunna
- Department of Neurosurgery, University of Missouri, Columbia, MO, USA
| | - Maxwell D. Gruber
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | | | - Zachary Taylor
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | | | - Fareed Jumah
- Department of Neurosurgery, University of Missouri, Columbia, MO, USA
| | - Periklis Godolias
- Swedish Medical Center, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Zachary Tataryn
- Swedish Medical Center, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Douglas Hollern
- Department of Orthopedic Surgery, University of Southern California, Los Angeles, CA, USA
| | - Rod Oskouian
- Swedish Medical Center, Swedish Neuroscience Institute, Seattle, WA, USA
| | - Jens R. Chapman
- Swedish Medical Center, Swedish Neuroscience Institute, Seattle, WA, USA
| |
Collapse
|
223
|
Fritzenschaft L, Boehm F, Rothenbacher D, Denkinger M, Dallmeier D. Association of blood biomarkers with frailty-A mapping review. Ageing Res Rev 2025; 109:102761. [PMID: 40318768 DOI: 10.1016/j.arr.2025.102761] [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: 12/31/2024] [Revised: 04/17/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Frailty describes a geriatric syndrome characterized by an increased vulnerability. Although a variety of potential blood-based biomarkers have been discussed for its characterization, a reliable protocol considering blood-based biomarkers for this purpose is still missing. However, a comprehensive overview on these biomarkers is necessary to understand potential molecular pathways to frailty. We, therefore, performed a mapping review to identify those blood-based biomarkers most consistently associated with frailty in community-dwelling older adults as well as possible analytical gaps according to the available literature. A proposed weighted correlation index (CI) describing the direction and consistency of the association considering the number of available publications as well as the size of the study populations was calculated for each biomarker. Overall, 72 manuscripts were critically reviewed reporting on a total of 82 biomarkers. The most consistent positive association with at least 3 articles addressing the respective biomarker in unadjusted and fully adjusted models was shown for interleukin 6 (IL-6), high-sensitivity C-reactive protein (hs-CRP), neopterin, white blood cells count, glycated hemoglobin A1c (HbA1c) and sex hormone binding-globuline (SHBG) with a CI ≥ 0.7, while for negative association hemoglobin, 25-hydroxy vitamin D, free testosterone in men and albumin with a CI ≤ -0.7 were identified.
Collapse
Affiliation(s)
- Lea Fritzenschaft
- Institute for Geriatric Research Ulm University Medical Center Ulm, Germany
| | - Felix Boehm
- Institute for Geriatric Research Ulm University Medical Center Ulm, Germany
| | | | - Michael Denkinger
- Institute for Geriatric Research Ulm University Medical Center Ulm, Germany; Department of Research on Ageing, AGAPLESION Bethesda Clinic Ulm, Ulm, Germany; Medical Faculty, Ulm University, Ulm, Germany
| | - Dhayana Dallmeier
- Institute for Geriatric Research Ulm University Medical Center Ulm, Germany; Department of Research on Ageing, AGAPLESION Bethesda Clinic Ulm, Ulm, Germany; Medical Faculty, Ulm University, Ulm, Germany; Department of Epidemiology, Boston University School of Public Health, Boston, USA.
| |
Collapse
|
224
|
Kaplan Z, Prezioso E, Jain A, Lavu H, Yeo CJ, Bowne WB, Nevler A. Clinical Implications of Mismatch Repair Deficiency in Pancreatic Ductal Adenocarcinoma. Cancer Med 2025; 14:e70960. [PMID: 40366030 PMCID: PMC12076359 DOI: 10.1002/cam4.70960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Pancreatic cancer is a highly aggressive and lethal disease, characterized by a limited response to chemotherapy and overall poor prognosis. Pancreatic cancers with a distinct mismatch repair deficiency, although relatively rare, have been shown to be associated with markedly better outcomes in comparison. Furthermore, whereas pancreatic cancers are generally unresponsive to current immunotherapy, this specific group of tumors has been shown to have a notable susceptibility to immune checkpoint inhibitors. AIMS In this review, we aim to summarize the relevant literature regarding mismatch-repair associated pancreatic cancers, the impacted biological mechanisms, and the resulting vulnerabilities for potential opportunistic immunotherapeutic treatment approaches. We will also review the current clinical studies assessing survival outcomes of mismatch repair deficient pancreatic cancers and ongoing clinical trials in this emerging field. RESULTS AND CONCLUSIONS Patients with dMMR/MSI-H pancreatic cancers harbor a distinct phenotype that has increased immune activation, greater responsiveness to immune checkpoint inhibitor therapy and better overall survival when compared to other pancreatic cancers. Although this molecular subtype makes up a small minority of cases, emerging data suggest immunotherapy may offer benefit to these patients.
Collapse
Affiliation(s)
- Zachary Kaplan
- Sidney Kimmel Medical CollegePhiladelphiaPennsylvaniaUSA
| | | | - Aditi Jain
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Harish Lavu
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Charles J. Yeo
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Wilbur B. Bowne
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| | - Avinoam Nevler
- Jefferson Pancreatic, Biliary, and Related Cancer CenterSidney Kimmel Cancer CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
225
|
Xiao Y, Jin W, Ju L, Fu J, Wang G, Yu M, Chen F, Qian K, Wang X, Zhang Y. Tracking single-cell evolution using clock-like chromatin accessibility loci. Nat Biotechnol 2025; 43:784-798. [PMID: 38724668 DOI: 10.1038/s41587-024-02241-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 04/10/2024] [Indexed: 05/18/2025]
Abstract
Single-cell chromatin accessibility sequencing (scATAC-seq) reconstructs developmental trajectory by phenotypic similarity. However, inferring the exact developmental trajectory is challenging. Previous studies showed age-associated DNA methylation (DNAm) changes in specific genomic regions, termed clock-like differential methylation loci (ClockDML). Age-associated DNAm could either result from or result in chromatin accessibility changes at ClockDML. As cells undergo mitosis, the heterogeneity of chromatin accessibility on clock-like loci is reduced, providing a measure of mitotic age. In this study, we developed a method, called EpiTrace, that counts the fraction of opened clock-like loci from scATAC-seq data to determine cell age and perform lineage tracing in various cell lineages and animal species. It shows concordance with known developmental hierarchies, correlates well with DNAm-based clocks and is complementary with mutation-based lineage tracing, RNA velocity and stemness predictions. Applying EpiTrace to scATAC-seq data reveals biological insights with clinically relevant implications, ranging from hematopoiesis, organ development, tumor biology and immunity to cortical gyrification.
Collapse
Affiliation(s)
- Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wan Jin
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Euler Technology, ZGC Life Sciences Park, Beijing, China
| | - Lingao Ju
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jie Fu
- Hong Kong University of Science and Technology, Hong Kong, China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mengxue Yu
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Fangjin Chen
- High Performance Computing Center, Peking-Tsinghua College of Life Sciences, Peking University, Beijing, China
| | - Kaiyu Qian
- Department of Biological Repositories, Human Genetic Resources Preservation Center of Hubei Province, Hubei Key Laboratory of Urological Diseases, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China.
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Yi Zhang
- Euler Technology, ZGC Life Sciences Park, Beijing, China.
| |
Collapse
|
226
|
Rüschoff J, Penner A, Ellis IO, Hammond MEH, Lebeau A, Osamura RY, Penault-Llorca F, Rojo F, Desai C, Moh A, Atkey N, Baenfer G, Scheel AH, D'Arrigo C, Schildhaus HU, Viale G. Global Study on the Accuracy of Human Epidermal Growth Factor Receptor 2-Low Diagnosis in Breast Cancer. Arch Pathol Lab Med 2025; 149:431-438. [PMID: 39111775 DOI: 10.5858/arpa.2024-0052-oa] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2024] [Indexed: 04/26/2025]
Abstract
CONTEXT.— Recently, a new type of antibody-drug conjugate, trastuzumab-deruxtecan (T-DXd), has been approved for the treatment of metastatic breast cancer with low level of human epidermal growth factor receptor 2 (HER2) gene expression. Thereby, eligibility relies on an accurate diagnosis of HER2-low status defined by immunohistochemistry IHC 1+/2+ with no gene amplification. OBJECTIVE.— To assess pathologists' accuracy and training efficacy in the diagnosis of HER2-low. DESIGN.— Agreement rates of HER2-low scoring in breast cancer tissue were assessed between expert consensus and real-world pathologists (n = 77 from 14 countries) before and after a specific 4-hour training program for HER2-low detection. Two assays were evaluated, the Ventana Pathway 4B5 CDx and the Dako HercepTest (polyclonal). Concordance of the pathologists with consensus score and efficacy of training were measured by Cohen κ, overall rater agreement, and receiver operating characteristic (ROC) curve statistics. RESULTS.— In the Ventana 4B5 HER2-low category, baseline agreement rates were >80% but <90%. Negative percentage agreement was improved from 80.6% to 91.1% by training. In the HER2-0 category, positive percentage agreement (74.6%) was the only parameter below the 80% benchmark but was significantly improved to 89.2% after training. Training efficacy was confirmed by ROC curve analysis, which shows improvement for the identification of HER2-0 and HER2-low cases. Finally, in-depth examination of cases with discordant HER2 status disclosed specific issues of HER2-low underscoring and overscoring. CONCLUSIONS.— The ability of pathologists to achieve acceptable diagnostic accuracy in identifying patients with HER2-low breast cancer could be enhanced by short-term training. Potential routes to improve the quality of HER2-low scoring in clinical practice have been identified.
Collapse
Affiliation(s)
- Josef Rüschoff
- From the Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany (Rüschoff, Penner, Baenfer, Schildhaus)
| | - Alexander Penner
- From the Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany (Rüschoff, Penner, Baenfer, Schildhaus)
| | - Ian O Ellis
- Translational Medical Sciences, School of Medicine, The University of Nottingham, Department of Histopathology, City Hospital Campus, Nottingham University Hospitals, Nottingham, United Kingdom (Ellis)
| | - M Elizabeth Hale Hammond
- Department of Pathology, Intermountain Healthcare and University of Utah School of Medicine, Salt Lake City (Hammond)
| | - Annette Lebeau
- University Medical Center Hamburg-Eppendorf, Institute of Pathology, Hamburg, Germany (Lebeau)
- Private Group Practice for Pathology, Lübeck, Germany (Lebeau)
| | - Robert Y Osamura
- Department of Diagnostic Pathology, Nippon Koukan Hospital, Kawasaki, Japan (Osamura)
| | - Fréderique Penault-Llorca
- Centre Jean Perrin, Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, Clermont Ferrand, France (Penault-Llorca)
| | - Federico Rojo
- Department of Pathology, IIS-Hospital Universitario Fundacion Jimenez Diaz-CIBERONC, Madrid, Spain (Rojo)
| | - Chirag Desai
- Daiichi Sankyo, Basking Ridge, New Jersey (Desai, Moh)
| | - Akira Moh
- Daiichi Sankyo, Basking Ridge, New Jersey (Desai, Moh)
| | - Neil Atkey
- Diaceutics plc, Dataworks at Kings Hall Life Sciences Park, Belfast, County Antrim, United Kingdom (Atkey)
| | - Gudrun Baenfer
- From the Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany (Rüschoff, Penner, Baenfer, Schildhaus)
| | - Andreas H Scheel
- Department of Pathology, University of Cologne, Cologne, Germany (Scheel)
| | - Corrado D'Arrigo
- Poundbury Cancer Institute, Dorchester, United Kingdom (D'Arrigo)
| | - Hans-Ulrich Schildhaus
- From the Discovery Life Sciences Biomarker Services GmbH, Kassel, Germany (Rüschoff, Penner, Baenfer, Schildhaus)
| | - Giuseppe Viale
- Department of Pathology, IEO European Institute of Oncology IRCCS Milan, Italy (Viale)
| |
Collapse
|
227
|
Harkos C, Hadjigeorgiou AG, Voutouri C, Kumar AS, Stylianopoulos T, Jain RK. Using mathematical modelling and AI to improve delivery and efficacy of therapies in cancer. Nat Rev Cancer 2025; 25:324-340. [PMID: 39972158 DOI: 10.1038/s41568-025-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
Mathematical modelling has proven to be a valuable tool in predicting the delivery and efficacy of molecular, antibody-based, nano and cellular therapy in solid tumours. Mathematical models based on our understanding of the biological processes at subcellular, cellular and tissue level are known as mechanistic models that, in turn, are divided into continuous and discrete models. Continuous models are further divided into lumped parameter models - for describing the temporal distribution of medicine in tumours and normal organs - and distributed parameter models - for studying the spatiotemporal distribution of therapy in tumours. Discrete models capture interactions at the cellular and subcellular levels. Collectively, these models are useful for optimizing the delivery and efficacy of molecular, nanoscale and cellular therapy in tumours by incorporating the biological characteristics of tumours, the physicochemical properties of drugs, the interactions among drugs, cancer cells and various components of the tumour microenvironment, and for enabling patient-specific predictions when combined with medical imaging. Artificial intelligence-based methods, such as machine learning, have ushered in a new era in oncology. These data-driven approaches complement mechanistic models and have immense potential for improving cancer detection, treatment and drug discovery. Here we review these diverse approaches and suggest ways to combine mechanistic and artificial intelligence-based models to further improve patient treatment outcomes.
Collapse
Affiliation(s)
- Constantinos Harkos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ashwin S Kumar
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
228
|
Li X, Tong K, Cheng T, Yan P, Xu H, Liu K, Xu R, Lu J, Yang Z, Wu H. Prediction of postoperative pancreatic fistula based on multi-sequence MR imaging. Eur J Radiol 2025; 186:112067. [PMID: 40147165 DOI: 10.1016/j.ejrad.2025.112067] [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: 12/31/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE To investigate the value of conventional MRI sequences in predicting the occurrence of postoperative pancreatic fistula (POPF) in patients undergoing pancreaticoduodenectomy (PD). METHODS A total of 122 patients from August 2019 to April 2023 were enrolled. All patients underwent pancreatic histological evaluation, including fibrosis, fat deposition, and acinar cell atrophy. The preoperative image features of pancreas were obtained, including morphological features, pancreas-muscle signal intensity ratio, pancreatic fat fraction and multi-phase enhancement features. The patients were divided into two groups according to whether pancreatic fistula occurred after operation. The related risk factors of pancreatic fistula, the correlation between imaging and pathological changes were analyzed, and the value of preoperative imaging in predicting pancreatic fistula was evaluated. RESULTS Of the 122 patients, 23(18.9 %) developed POPF. Pathological score showed that there was a significant difference in pancreatic fat deposition between the two groups (P = 0.006), the fat deposition score was higher in the POPF group. Pancreatic fat deposition was the only independent risk factor for POPF(OR,1.933; P = 0.018). MRI showed that proton density fat fraction(PDFF) (P = 0.001), pancreas-to-aorta signal intensity ratio(P-A SI ratio) of equilibrium phase(P = 0.023) and delay phase(P = 0.020) had significant differences. PDFF was positively correlated with fat deposition(r = 0.404, P < 0.001), P-A SI ratio of equilibrium phase and delay phase were positively correlated with fibrosis(r = 0.313, P = 0.002; r = 0.315, P = 0.002, respectively). ROC analysis showed that PDFF had the best efficacy in predicting postoperative pancreatic fistula (AUROC = 0.810), better than P-A SI ratio of equilibrium phase(AUROC = 0.752) and delayed phase(AUROC = 0.766). CONCLUSIONS Pancreatic fat deposition is a high risk factor for POPF, PDFF can reflect fat deposition and predict POPF.
Collapse
Affiliation(s)
- Xiaoyang Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Kuinan Tong
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Tianxin Cheng
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Piao Yan
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Hui Xu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Kun Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Rui Xu
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Jun Lu
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China
| | - Zhenghan Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China.
| | - Hongwei Wu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Yongan Road 95, West District, Beijing 100050, China.
| |
Collapse
|
229
|
Yost KE, Zhao Y, Hung KL, Zhu K, Xu D, Corces MR, Shams S, Louie BH, Sarmashghi S, Sundaram L, Luebeck J, Clarke S, Doane AS, Granja JM, Choudhry H, Imieliński M, Cherniack AD, Khurana E, Bafna V, Felau I, Zenklusen JC, Laird PW, Curtis C, Greenleaf WJ, Chang HY. Three-dimensional genome landscape of primary human cancers. Nat Genet 2025; 57:1189-1200. [PMID: 40355593 DOI: 10.1038/s41588-025-02188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/02/2025] [Indexed: 05/14/2025]
Abstract
Genome conformation underlies transcriptional regulation by distal enhancers, and genomic rearrangements in cancer can alter critical regulatory interactions. Here we profiled the three-dimensional genome architecture and enhancer connectome of 69 tumor samples spanning 15 primary human cancer types from The Cancer Genome Atlas. We discovered the following three archetypes of enhancer usage for over 100 oncogenes across human cancers: static, selective gain or dynamic rewiring. Integrative analyses revealed the enhancer landscape of noncancer cells in the tumor microenvironment for genes related to immune escape. Deep whole-genome sequencing and enhancer connectome mapping provided accurate detection and validation of diverse structural variants across cancer genomes and revealed distinct enhancer rewiring consequences from noncoding point mutations, genomic inversions, translocations and focal amplifications. Extrachromosomal DNA promoted more extensive enhancer rewiring among several types of focal amplification mechanisms. These results suggest a systematic approach to understanding genome topology in cancer etiology and therapy.
Collapse
Affiliation(s)
- Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Yanding Zhao
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - King L Hung
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Pathos AI, Chicago, IL, USA
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shadi Shams
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan H Louie
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Laksshman Sundaram
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
- NVIDIA Bio Research, NVIDIA, Santa Clara, CA, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Stanley Clarke
- Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York City, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Ashley S Doane
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Jeffrey M Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marcin Imieliński
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Andrew D Cherniack
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York City, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Ina Felau
- National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Christina Curtis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
230
|
Uno R, Zen Y, Tanaka T, Toyama H, Fukumoto T, Sofue K, Itoh T. A Potential Case of Tubulocystic Ductal Carcinoma of the Pancreas. Pathol Int 2025; 75:243-246. [PMID: 40243254 DOI: 10.1111/pin.70008] [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: 10/01/2024] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025]
Abstract
We present a case of pancreatic ductal carcinoma with a microcystic appearance. A 64-year-old woman was found to have a pancreatic mass during a routine medical checkup. The tumor was well-circumscribed and multicystic; thus, she was followed up for suspected serous cystadenoma. However, the tumor gradually enlarged in the following 2.5 years; subsequently, she underwent a Whipple procedure. Grossly, the cut surface of the tumor was honeycomb-like with small cysts. Histologically, the cysts were ductal structures lined by a relatively bland, cuboidal or columnar epithelium with mildly enlarged nuclei. No intracytoplasmic mucus was observed. The presence of stromal invasion confirmed the diagnosis of ductal carcinoma. KRAS was wild type. Postoperative course was uneventful, with no recurrence to date (followup period: 5.5 years postsurgery). The present case did not meet any known subtypes of pancreatic ductal carcinoma. The tumor resembled a large duct variant, which typically shows a microcystic appearance. However, unlike the present case, the large duct type usually consists of mucus-rich neoplastic cells. A recent study on cholangiocarcinoma proposed a novel tubulocystic subtype characterized by microcystic neoplastic glands and adenofibromatous stroma, which is morphologically similar to the present case. The present case may correspond to a pancreatic counterpart of tubulocystic cholangiocarcinoma.
Collapse
Affiliation(s)
- Rena Uno
- Department of Diagnostic Pathology, Hyogo Cancer Center, Kita Oji-cho, Akashi City, Hyogo Prefecture, Japan
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe City, Hyogo Prefecture, Japan
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Tomonori Tanaka
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe City, Hyogo Prefecture, Japan
| | - Hirochika Toyama
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Hospital, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Takumi Fukumoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Hospital, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe City, Hyogo Prefecture, Japan
| | - Tomoo Itoh
- Department of Diagnostic Pathology, Kobe University Hospital, Kobe City, Hyogo Prefecture, Japan
| |
Collapse
|
231
|
Di Gennaro F, Cotugno S, Guido G, Cavallin F, Papagni R, De Vita E, Manco Cesari G, Di Gregorio S, Giliberti V, Cassano D, Metrangolo G, Amendolara A, Francabandiera A, Fiorella M, Lobalsamo V, Brindicci G, Santoro CR, Ronga L, Lattanzio R, De Iaco G, Saracino A. Impact of a TB Team on TB Outcomes: A 2016-2024 Pre-Post Study From a Referral Center in Southern Italy. Open Forum Infect Dis 2025; 12:ofaf258. [PMID: 40376187 PMCID: PMC12079650 DOI: 10.1093/ofid/ofaf258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
Background Tuberculosis (TB) remains a major global public health challenge, requiring innovative management strategies to improve patient outcomes and reduce disease burden. Despite advancements in diagnostics and treatment, TB continues to cause significant morbidity and mortality worldwide, particularly among vulnerable populations such as migrants and individuals with limited healthcare access. The World Health Organization has emphasized the need for patient-centered approaches, including the establishment of TB-dedicated teams, to enhance care coordination and improve outcomes. However, real-world evidence on their effectiveness remains limited. Methods We conducted a retrospective before-after study to evaluate the impact of a structured TB-dedicated team implemented at a referral hospital in Southern Italy between 2016 and 2024. Prior to 2020, TB care was unstructured, with patients being followed by different physicians without a standardized system. Between 2021 and 2024, a dedicated TB team was established, comprising 2 infectious disease specialists, infectious disease residents, and a senior professor overseeing scientific activities. This new model introduced key interventions, including a dedicated outpatient reservation system, directly observed therapy, structured follow-up, specialist networking, systematic data collection, and targeted counseling on risk behaviors and comorbidities. Primary outcomes included treatment success rates, hospital length of stay, incidence of adverse events, and patient retention in follow-up programs. All tests were 2-sided and a P-value <.05 was considered statistically significant. Statistical analysis was carried out using R 4.4. Results A total of 269 TB patients were analyzed (117 pre- and 152 postimplementation). The introduction of the TB team was associated with a significant reduction in loss to follow-up (41.9% to 3.7%; P < .0001; odds ratio, 0.04; 95% confidence interval, .01-.12) and incomplete treatments (41.9% to 12.0; P < .0001; odds ratio, 0.13; 95% confidence interval, .06-.29). Median hospital stay decreased from 28 to 14 days (P < .0001), whereas adverse events remained comparable (32.5% vs 29.6%, P = .71). Conclusions The implementation of a TB-dedicated team significantly improved TB management by enhancing treatment adherence, reducing hospitalization, and preventing loss to follow-up. Given the public health impact of TB, structured care models should be prioritized to optimize patient outcomes and strengthen health system efficiency.
Collapse
Affiliation(s)
- Francesco Di Gennaro
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Sergio Cotugno
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Giacomo Guido
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | | | - Roberta Papagni
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Elda De Vita
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Giorgia Manco Cesari
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Stefano Di Gregorio
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Giliberti
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Domenica Cassano
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Giuliana Metrangolo
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Angela Amendolara
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Alessandro Francabandiera
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Monica Fiorella
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Vittoria Lobalsamo
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Gaetano Brindicci
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Carmen Rita Santoro
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Ronga
- Microbiology and Virology Unit, University of Bari, University Hospital Policlinico, Bari, Italy
| | - Rossana Lattanzio
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Giuseppina De Iaco
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Saracino
- Department of Precision and Regenerative Medicine and Jonian Area (DiMePreJ), Clinic of Infectious Diseases, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
232
|
Prokopidis K, Testa GD, Giannaki CD, Stavrinou P, Kelaiditi E, Hoogendijk EO, Veronese N. Prognostic and Associative Significance of Malnutrition in Sarcopenia: A Systematic Review and Meta-Analysis. Adv Nutr 2025; 16:100428. [PMID: 40222723 PMCID: PMC12099873 DOI: 10.1016/j.advnut.2025.100428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025] Open
Abstract
Malnutrition is a common phenomenon, particularly in those at an increased risk of muscle mass and function losses. In this systematic review and meta-analysis, we aimed to explore the association of malnutrition with sarcopenia in middle-aged and older adults and the prognostic association of malnutrition and sarcopenia compared with sarcopenia alone on all-cause mortality. PubMed, Scopus, Web of Science, and Cochrane Library were searched from inception until January 2024. A meta-analysis using a random-effect model was employed, utilizing the Mini Nutritional Assessment malnutrition tool as a continuous and categorical variable. The study protocol was registered in the International Prospective Register of Systematic Reviews (CRD42024501521). Malnutrition was significantly associated with a greater risk of sarcopenia [continuous: k = 12, odds ratio (OR): 1.38, 95% confidence interval (CI): 1.18, 1.61, I2 = 94.8%, P < 0.01; categorical: k = 37, OR: 2.99, 95% CI: 2.26, 3.96, I2 = 78.3%, P < 0.01]. Sarcopenia and malnutrition were associated with a higher risk of mortality compared with sarcopenia alone (k = 5, hazard ratio: 4.04, 95% CI: 1.36, 11.94, I2 = 92.8%, P < 0.01). Metaregression showed age, sex, and number of adjustments did not explain heterogeneity among studies. The included studies had a moderate risk of bias. Malnutrition is associated with higher odds of sarcopenia and their combined presence is a better predictor of all-cause mortality compared with sarcopenia alone, further highlighting the importance of applying interventions to counteract these two closely related phenomena.
Collapse
Affiliation(s)
- Konstantinos Prokopidis
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Giuseppe Dario Testa
- Department of Geriatric and Intensive Care Medicine, Careggi Hospital, University of Florence, Florence, Italy
| | - Christoforos D Giannaki
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Pinelopi Stavrinou
- Department of Life Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Eirini Kelaiditi
- Faculty of Sport, Allied Health and Performance Science, St Mary's University, Twickenham, United Kingdom
| | - Emiel O Hoogendijk
- Department of General Practice, Amsterdam Public Health research institute, VU University Medical Center, Amsterdam, The Netherlands; Department of Epidemiology & Data Science, Amsterdam Public Health research institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Nicola Veronese
- Department of Health Promotion, Mother Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| |
Collapse
|
233
|
Luchini C, PathologyOutlines.com Review Committee, PathologyOutlines.com Contributors, Pernick N. Benign Tumors and Tumor-Like Conditions of Ampulla and Small Intestine: The PathologyOutlines.com Review. Int J Surg Pathol 2025; 33:528-539. [PMID: 39377108 DOI: 10.1177/10668969241283748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Small intestine and ampulla can be involved in a heterogeneous group of benign tumors and tumor-like diseases, potentially mimicking malignant neoplasms. In-depth knowledge of them is critical for practicing pathologists since they represent potential pitfalls in routine diagnostic activity. Such conditions include the following: (1) Brunner gland lesions, including Brunner gland hyperplasia and hamartoma; (2) polyps, such as adenoma, hyperplastic, hamartomatous, and inflammatory; (3) mesenchymal proliferations encompassing inflammatory fibroid polyp, inflammatory myofibroblastic tumor, leiomyomas, lipomas, and lipomatosis of the ileocecal valve; (4) fibrosis-associated diseases, such as idiopathic retroperitoneal fibrosis, reactive nodular fibrous pseudotumor, and sclerosing peritonitis; (5) disorders of lymphatic vessels, including lymphangiectasia and lymphangiomas; and (6) other rare conditions/miscellanea, such as enteritis cystica profunda, intussusception, ischemia, and pneumatosis cystoides intestinalis. This review, inspired by the content of the PathologyOutlines website (https://www.pathologyoutlines.com), aims to provide a reference point in this complex scenario, summarizing the essential histopathological features of all these entities for better addressing routine practice and differential diagnoses.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, and ARC-NET Research Center, University of Verona, Verona, Italy
| | | | | | | |
Collapse
|
234
|
Miller CH, Vemuri A, Lengyel E, Lastra RR. Adaptive immune response and PD-1/ PD-L1 status in chemotherapy treated high grade serous carcinoma is dependent on chemotherapy response score. Hum Pathol 2025; 159:105800. [PMID: 40389122 DOI: 10.1016/j.humpath.2025.105800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Platinum-based chemotherapy and debulking surgery is the standard of care for patients with advanced tubo-ovarian high-grade serous carcinoma (HGSC). The chemotherapy response scoring (CRS) system is a histopathologic scoring system developed to measure response to neoadjuvant chemotherapy with prognostic implications. Omental samples with high CRS have greater inflammatory cell infiltrates, but the immunophenotype of infiltrating immune cells and PD-L1 expression of the residual tumor has not been well-defined. DESIGN Twenty cases of patients with FIGO stage IIIA to IIIC HGSC undergoing interval debulking after receiving 3-4 rounds of chemotherapy were selected. 6/20 cases of omental samples were graded as CRS 1, 7/20 were graded CRS 2, and 7/20 were graded CRS 3. The following immunohistochemical stains were performed: CD8, CD4, Foxp3, PD1, and PD-L1. The total number of tumor-infiltrating lymphocytes was recorded, and each case was given a PD-L1 combined positive score (CPS) and tumor proportion score (TPS). RESULTS There was a significantly greater number of CD8+ T cells, PD-1+ T cells, CD4+ T cells, and Foxp3+ T cells in CRS 3-scored cases compared to CRS 1 scored cases (p-values: 0.0018, 0.0224, 0.0071, and 0.0136, respectively). CRS 3-scored cases had a greater PD-L1 CPS (CRS 3 CPS 13 ± 8.2 versus CRS 1 CPS 0 ± 0; p-value: 0.0485). CONCLUSIONS Tubo-ovarian high-grade serous carcinoma with greater response to neoadjuvant treatment have significantly greater T cell infiltrate and greater PD-L1 combined positive score, highlighting a potential role of the CRS as a predictive biomarker for immune checkpoint blockade therapy.
Collapse
Affiliation(s)
| | - Anusha Vemuri
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/ Section of Gynecologic Oncology, University of Chicago, IL, USA
| | - Ricardo R Lastra
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
235
|
Weeber L, Su N, Faggion CM. Characteristics of search methods in dental meta-research studies: a methodological study. J Clin Epidemiol 2025; 181:111693. [PMID: 39892521 DOI: 10.1016/j.jclinepi.2025.111693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND AND OBJECTIVE Meta-research studies, defined as, "research on research," should transparently report search methods used to identify the assessed research. Currently, there is no published evaluation of search methods reporting in meta-research studies. The aim of this study was to assess the characteristics of search methods in dental meta-research studies and to identify factors associated with the completeness of the reported search strategies. METHODS With a focus on the assessment of reporting quality and methodological quality, we searched in the Web of Science (WoS) Core Collection database for dental meta-research studies published from the database's inception to February 13, 2024. The extracted data included the examined meta-research studies, characteristics of their authors and journals, and search methods reporting of the examined studies. Logistic regression models were applied to examine the associations between relevant variables and search strategy reporting completeness. RESULTS The search generated 3774 documents, and 224 meta-research studies were included in the final analysis. Nearly all studies (99.6%) disclosed their general search methods, but only 130 studies (58%) provided both keywords and Boolean operators. Regression analyses indicated that meta-research studies published more recently, with prospective registration, with a shorter time between the searches and publication, a lack of language restrictions, and librarian involvement were more likely to report a more complete search strategy. CONCLUSION The results highlight the importance of unrestricted language searches, structured methodologies, and librarian support in improving the quality and transparency of reporting search strategies in dental meta-research.
Collapse
Affiliation(s)
- Leonie Weeber
- Department of Periodontology and Operative Dentistry, University Hospital Münster, Münster, Germany
| | - Naichuan Su
- Department of Oral Public Health, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam 1081 LA, The Netherlands
| | - Clovis Mariano Faggion
- Department of Periodontology and Operative Dentistry, University Hospital Münster, Münster, Germany.
| |
Collapse
|
236
|
Liu Y, Peng J, Zhao Y, Wang W. Emerging pathological diagnostic strategies for solid pseudopapillary neoplasm of the pancreas: insights from omics and innovative techniques. J Pathol Clin Res 2025; 11:e70029. [PMID: 40312910 PMCID: PMC12046068 DOI: 10.1002/2056-4538.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/21/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
Solid pseudopapillary neoplasm (SPN) of the pancreas is a rare, low-grade malignant tumor, representing 0.9-2.7% of all exocrine pancreatic tumors. SPN patients generally have a favorable prognosis with a 5-year survival rate exceeding 95% following complete surgical resection. Accurate diagnosis is crucial to avoid unnecessary treatments. Currently, SPN diagnosis relies on imaging techniques such as CT and MRI, along with immunohistochemical analysis of biopsy and resection samples. The main challenge in diagnosis is the potential inability to accurately identify recurrent or metastatic SPN, as well as 'malignant' SPN, due to the lack of specific biomarkers. Advances in high-throughput omics technologies, including genomics, transcriptomics, proteomics and metabolomics, have opened new avenues for identifying novel biomarkers for SPN. Additional, liquid biopsy techniques have enabled more comprehensive analysis of biosamples such as pancreatic cyst fluid, offering promising prospects for preoperative diagnosis. This review highlights recent research on SPN diagnosis, focusing on immunohistochemical markers, tissue sampling methods and the potential of omics approaches. It also discusses the challenges and opportunities in improving diagnostic accuracy, particularly for high-grade and metastatic SPNs.
Collapse
Affiliation(s)
- Yuanhao Liu
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Junya Peng
- Institute of Clinical MedicinePeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
| | - Yupei Zhao
- State Key Laboratory of Complex, Severe, and Rare DiseasesPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of General SurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Department of Basic Medical SciencesSchool of Medicine, Tsinghua UniversityBeijingPR China
- Peking University‐Tsinghua Center for Life SciencesBeijingPR China
| | - Wenze Wang
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical CollegeBeijingPR China
- Molecular Pathology Research Center, Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPR China
| |
Collapse
|
237
|
Elleuch D, Chen Y, Luo Q, Palaniyappan L. Speaking of yourself: A meta-analysis of 80 years of research on pronoun use in schizophrenia. Schizophr Res 2025; 279:22-30. [PMID: 40157253 DOI: 10.1016/j.schres.2025.03.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/16/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
People with schizophrenia experience significant language disturbances that profoundly affect their everyday social interactions. Given its relevance to the referential function of language, aberrations in pronoun use are of particular interest in the study of schizophrenia. This systematic review and meta-analysis, adhering to PRISMA guidelines, examines the frequency of pronoun use in schizophrenia. PubMed, PsycINFO, Scopus, Google Scholar, and Web of Science were searched up to May 1, 2024. All studies analyzing pronoun frequency in various spoken language contexts in schizophrenia were included. Bias was assessed using a modified Newcastle-Ottawa Scale. A Bayesian meta-analysis with model averaging estimated effect sizes and moderating factors. 13 studies with n = 917 unique participants and 13 case-control contrasts were included. 37.9 % of patient samples were women, with a weighted mean (SD) age of 34.45 (9.72) years. 53.85 % of the studies were in languages other than English. We report a medium-sized effect for first-person pronoun impairment in schizophrenia (model-averaged d = 0.89, 95 % CrI (0.44, 1.33)). There was significant heterogeneity moderated by age. Evidence for publication bias was weak, with a strong support for first-person pronoun impairment after accounting for bias and heterogeneity. There was a small reduction of inter-individual variability in first-person pronoun use in patients compared to healthy controls (lnCVR = -0.12, 95 % CrI [-0.35, -0.13]). While all pronoun use was also high in patients, this was not robust due to heterogeneity and publication bias. Individuals with schizophrenia excessively use first-person pronouns. This may be a marker of a disturbed sense of self in this illness.
Collapse
Affiliation(s)
- Dalia Elleuch
- Higher School of Health Sciences and Techniques of Sfax, University of Sfax, Tunisia; Laboratory of Neuroscience, Faculty of Medicine of Sfax, University of Sfax, Tunisia
| | - Yinhan Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Research Institute of Intelligent Complex Systems, Fudan University, Shanghai 200433, China
| | - Qiang Luo
- Institute of Science and Technology for Brain-Inspired Intelligence, Research Institute of Intelligent Complex Systems, Fudan University, Shanghai 200433, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200433, China
| | - Lena Palaniyappan
- Douglas Mental Health University Institute, Department of Psychiatry, McGill University, Quebec, Canada; Robarts Research Institute & Lawson Health Research Institute, London, Ontario, Canada; Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
238
|
Duhn J, von Fritsch L, Bolm L, Braun R, Honselmann K, Litkevych S, Kist M, Deichmann S, Tol KKV, Franke B, Reinwald F, Sackmann A, Holleczek B, Krauß A, Klinkhammer-Schalke M, Zeissig SR, Keck T, Wellner UF, Abdalla TSA. Perioperative and oncologic outcomes after total pancreatectomy and pancreatoduodenectomy for pancreatic head adenocarcinoma-A propensity score-matched analysis from the German Cancer Registry Group. Surgery 2025; 181:109292. [PMID: 40101369 DOI: 10.1016/j.surg.2025.109292] [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: 11/13/2024] [Revised: 01/08/2025] [Accepted: 02/01/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND To compare perioperative morbidity and mortality in patients receiving pancreatoduodenectomy or total pancreatectomy for pancreatic head adenocarcinoma using German Cancer Registry data. METHODS Anonymized pooled data were retrieved from regional cancer registries participating in the German Cancer Registry Group of the Association of German Tumor Centers. Included were patients diagnosed with pancreatic head adenocarcinoma since 2016, receiving curative intent pancreatoduodenectomy or total pancreatectomy. Patients were propensity-score matched according to age, sex, and histopathology. Primary endpoints were 30- and 90-day postoperative mortality. Secondary endpoints were administration of adjuvant chemotherapy, long-term survival, and patterns of cancer recurrence. The data were analyzed using R. RESULTS In total, 756 patients per treatment group were matched for further analyses. R0-resection rate was comparable between pancreatoduodenectomy and total pancreatectomy (69.6 vs 73.4%, P = .154). The 30-day (9.5 vs 4.8%, P < .001) and 90-day postoperative mortality (18.0 vs 11.0%, P < .001) rates were significantly lower after pancreatoduodenectomy compared with total pancreatectomy. After pancreatoduodenectomy, more patients received adjuvant chemotherapy (43.6 vs 53.3%, P < .001) and time to adjuvant chemotherapy was shorter (60.1 vs 52.7 days, P = .002) compared with total pancreatectomy. Long-term overall survival was worse after total pancreatectomy (P < .001), also in patients receiving adjuvant chemotherapy (P = .019). The sites of recurrence were comparable between both groups (P = .274). CONCLUSION The results of this study show greater perioperative morbidity and mortality after total pancreatectomy compared with pancreatoduodenectomy for pancreatic head malignancy. Also, long-term survival was worse after total pancreatectomy. These results emphasize the role of pancreatoduodenectomy as a standard surgical procedure for pancreatic head adenocarcinoma and suggest that total pancreatectomy should only be performed in selected patients.
Collapse
Affiliation(s)
- Jannis Duhn
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Lennart von Fritsch
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Louisa Bolm
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Rüdiger Braun
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kim Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Stanislav Litkevych
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Markus Kist
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Steffen Deichmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Kees Kleihues-van Tol
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Bianca Franke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Fabian Reinwald
- Cancer Registry of Rhineland-Palatinate in the Institute for Digital Health Data, Mainz, Germany
| | - Andrea Sackmann
- Hessian Cancer Registry, Hessian Office for Health and Care, Frankfurt, Germany
| | | | - Anna Krauß
- Cancer Registry Mecklenburg-Western Pomerania, Greifswald, Germany
| | - Monika Klinkhammer-Schalke
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany
| | - Sylke R Zeissig
- Network for Care, Quality and Research in Oncology, German Cancer Registry Group of the Association of German Tumor Centers (ADT), Berlin, Germany; Institute of Clinical Epidemiology and Biometry (ICE-B), University of Würzburg, Würzburg, Germany
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany.
| | - Ulrich F Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | - Thaer S A Abdalla
- Department of Surgery, University Medical Center Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| |
Collapse
|
239
|
Tan CHN, Yeo B, Vasanwala RF, Sultana R, Lee JH, Chan D. Vitamin D Deficiency and Clinical Outcomes in Critically Ill Pediatric Patients: A Systematic Review and Meta-Analysis. J Endocr Soc 2025; 9:bvaf053. [PMID: 40242208 PMCID: PMC12001026 DOI: 10.1210/jendso/bvaf053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Indexed: 04/18/2025] Open
Abstract
Context Vitamin D deficiency (VDD) is common in paediatric populations, and its relationship with critical care outcomes warrants further investigation. Objective The aim is to examine the association between VDD and clinical outcomes in children admitted to the Pediatric Intensive Care Unit (PICU). Methods This systematic review and meta-analysis investigated the impact of VDD on clinical outcomes in PICU patients. A comprehensive search of Embase, Web of Science, PubMed, and Cochrane databases was conducted. Our primary outcomes were mortality and sepsis incidence, while secondary outcomes included length of stay (LOS), need for inotropic support, and need for and duration of mechanical ventilation. Eligible studies included infants and children aged 1 month to 18 years admitted to the PICU, with baseline 25-hydroxyvitamin D levels measured on admission. Two independent reviewers screened studies, extracted data, and assessed quality. Pooled estimates were obtained using a random-effects model. Results Out of 2298 screened studies, 27 met the inclusion criteria, comprising 4682 patients. VDD was defined as 25-hydroxyvitamin D levels <20 ng/mL and <30 ng/mL in 22 and 5 studies, respectively. VDD was associated with increased mortality (odds ratio [OR] 2.05, 95% CI 1.21-3.48) and a greater need for inotropic support (OR 2.02, 95% CI 1.43-2.85) than children with vitamin D sufficiency (VDS). No differences were observed between VDD and VDS groups in terms of sepsis incidence postadmission, LOS, or the need for and duration of mechanical ventilation. Conclusion VDD in critically ill pediatric patients was associated with increased mortality and higher need for inotropic support. Further research is warranted to evaluate the potential benefits of vitamin D supplementation in this high-risk population.
Collapse
Affiliation(s)
- Chai-Hoon Nowel Tan
- Department of Pediatrics, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Bernita Yeo
- Department of Pediatrics, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Rashida Farhad Vasanwala
- Endocrinology Service, Department of Pediatric Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
- Pediatrics Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Rehena Sultana
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jan Hau Lee
- Pediatrics Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
- Pediatric Intensive Care, Department of Pediatric Subspecialties, KK Women's and Children's Hospital, Singapore 229899, Singapore
| | - Daniel Chan
- Endocrinology Service, Department of Pediatric Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore
- Pediatrics Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| |
Collapse
|
240
|
Rueff J, Conde J, Castro G. Exploring the link between low germline mutational load and low breast cancer incidence: Lessons from the Xavante Indians. Transl Oncol 2025; 55:102356. [PMID: 40086325 PMCID: PMC11957485 DOI: 10.1016/j.tranon.2025.102356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
The study of cancer, its initiation, and its mechanisms of progression has been a focal point in science for more than a century. Despite controversies among scientists, there is a growing consensus to determine the moment when a cell gains the capacity to be transformed and whether this mechanism is to be attributed to germinal or somatic events, or possibly both. The case of the Xavante Indians is a beacon for this journey, pointing toward the importance of genetic diversity in shaping our approach to cancer research and treatment. As we incorporated these lessons into clinical practice, we embarked on a new era of personalized preventative healthcare strategies against cancer. Based on recent data, we comment on the low germinal mutational load and low cancer incidence. Statistical analyses reveal a significantly lower mutation burden in Xavante women compared to global populations (p < 0.0001), including rare deleterious variants in cancer-associated genes. Additionally, polygenic risk scores (PRS) for breast cancer are markedly lower in Xavante (mean PRS ∼35) compared to TCGA cohorts (∼80-90) (p < 0.0001). The absence of breast cancer cases in Xavante is statistically significant when compared to expected rates (p < 0.001), reinforcing the hypothesis of a protective genetic landscape.
Collapse
Affiliation(s)
- José Rueff
- NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, Portugal; ToxOmics, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.
| | - João Conde
- NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, Portugal; ToxOmics, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.
| | - Guilherme Castro
- Molecular Cancer Research Center, P.O. Box 1559, Woking, GU22 2WN, UK.
| |
Collapse
|
241
|
Jamouss KT, Damanakis AI, Cornwell AC, Jongepier M, Trujillo MA, Pflüger MJ, Kawalerski R, Maalouf A, Hirose K, Datta S, Sipes A, Pedro BA, Gudmundsson E, Assarzadegan N, Engle L, Scharpf RB, Kawamoto S, Thompson ED, Wood LD. Tumor immune microenvironment alterations associated with progression in human intraductal papillary mucinous neoplasms. J Pathol 2025; 266:40-50. [PMID: 40001347 DOI: 10.1002/path.6402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 11/26/2024] [Accepted: 01/08/2025] [Indexed: 02/27/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge due to late-stage diagnoses. To improve patient outcomes, early intervention in precursor lesions such as intraductal papillary mucinous neoplasm (IPMN) is crucial. However, early intervention must be balanced against overtreatment of low-risk lesions that are unlikely to progress, underscoring the need to better understand molecular alterations in neoplastic cells and changes in the tumor microenvironment (TME) that drive the progression of IPMNs. In this study, we characterized alterations in the TME of IPMNs as they progressed to high-grade dysplasia, using immunohistochemistry to quantify immune cell density and activation status in more than 100 well-characterized human IPMN samples. Analyses revealed progression to a more immunosuppressive TME in high-grade IPMN compared with low-grade IPMN, characterized by elevated expression of immune checkpoint molecules (PD-L1, TIM3, VISTA), increased density of macrophages, and decreased density of cytotoxic T cells. Intriguingly, the alterations in macrophages were limited to focal regions of high-grade dysplasia, while T-cell alterations affected the entire IPMN. Additionally, elevated VISTA expression was associated with poorer clinical outcome after IPMN resection in an independent cohort. These findings provide important insights into the interplay between the immune microenvironment and IPMN progression, highlighting potential targets to modify the TME for cancer interception. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Kevin T Jamouss
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander Ioannis Damanakis
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abigail C Cornwell
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martine Jongepier
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maria A Trujillo
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Johannes Pflüger
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Graduate School of Life Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ryan Kawalerski
- Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandre Maalouf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katsuya Hirose
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shalini Datta
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Abigail Sipes
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian A Pedro
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emma Gudmundsson
- Department of Physiology, University of Maryland, Baltimore, MD, USA
| | - Naziheh Assarzadegan
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Logan Engle
- Bloomberg Kimmel Institute, Tumor Microenvironment Technology Development Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Satomi Kawamoto
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth D Thompson
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Laura D Wood
- Department of Pathology, Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
242
|
Bruhm DC, Vulpescu NA, Foda ZH, Phallen J, Scharpf RB, Velculescu VE. Genomic and fragmentomic landscapes of cell-free DNA for early cancer detection. Nat Rev Cancer 2025; 25:341-358. [PMID: 40038442 DOI: 10.1038/s41568-025-00795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
Genomic analyses of cell-free DNA (cfDNA) in plasma are enabling noninvasive blood-based biomarker approaches to cancer detection and disease monitoring. Current approaches for identification of circulating tumour DNA typically use targeted tumour-specific mutations or methylation analyses. An emerging approach is based on the recognition of altered genome-wide cfDNA fragmentation in patients with cancer. Recent studies have revealed a multitude of characteristics that can affect the compendium of cfDNA fragments across the genome, collectively called the 'cfDNA fragmentome'. These changes result from genomic, epigenomic, transcriptomic and chromatin states of an individual and affect the size, position, coverage, mutation, structural and methylation characteristics of cfDNA. Identifying and monitoring these changes has the potential to improve early detection of cancer, especially using highly sensitive multi-feature machine learning approaches that would be amenable to broad use in populations at increased risk. This Review highlights the rapidly evolving field of genome-wide analyses of cfDNA characteristics, their comparison to existing cfDNA methods, and recent related innovations at the intersection of large-scale sequencing and artificial intelligence. As the breadth of clinical applications of cfDNA fragmentome methods have enormous public health implications for cancer screening and personalized approaches for clinical management of patients with cancer, we outline the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Daniel C Bruhm
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicholas A Vulpescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachariah H Foda
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
243
|
Lopes MP, Ahmed S, Beaman L, Stubbs B, Campbell IC, Schmidt U, Robinson L. Bone Fracture History in Women With First Episode or With Persistent Anorexia Nervosa. EUROPEAN EATING DISORDERS REVIEW 2025; 33:447-459. [PMID: 39572404 PMCID: PMC11965545 DOI: 10.1002/erv.3153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/23/2024] [Accepted: 10/31/2024] [Indexed: 04/04/2025]
Abstract
OBJECTIVE To investigate fractures history in women with first episode anorexia nervosa (AN) (FE-AN: ≤ 3 years duration) and those with persistent AN (P-AN: ≥ 7 years), compared to healthy controls (HC). METHOD One hundred nineteen women (FE-AN = 49, P-AN = 46 and HC = 24) completed online questionnaires on eating disorders symptoms, their menstrual and their fracture history. RESULTS Average illness duration was 1.9 years (SD = 0.8) in FE-AN and 15.3 years (SD = 8.5) in P-AN. Lifetime history of all fractures, including stress fractures, was higher in AN groups (FE-AN = 33.3%; P-AN = 37.8%) than in HC (4.2%, p < 0.001). P-AN participants were 13.4 times more likely to report a fracture compared to HC, irrespective of age, whereas F-AN participants were 10.3 times more likely. In P-AN, higher BMI, shorter duration of amenorrhoea and history of pregnancy were inversely associated with fracture number. CONCLUSIONS There is an increased risk of bone fracture even in the early stages of AN. This could be related to a time lapse between the initial symptoms of AN and formal diagnosis. This suggests guidelines recommending bone screening after 2-years of persistent low weight for adults should be revisited, and the risk of bone problems should be part of the dialogue between clinicians, patients and carers at the earliest opportunity.
Collapse
Affiliation(s)
- Mariana P. Lopes
- Department of Psychological MedicineCentre for Research in Eating and Weight DisordersInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- Nutrition DepartmentSchool of Public Health University of São PauloSão PauloBrazil
| | - Sana Ahmed
- Social, Genetic and Developmental Psychiatry (SGDP) CentreInstitute of PsychiatryPsychology & NeuroscienceKing's College LondonLondonUK
- Syed Babar Ali School of Science and EngineeringLahore University of Management SciencesLahorePakistan
| | - Lily Beaman
- Psychology and NeuroscienceInstitute of PsychiatryKings College LondonLondonUK
| | - Brendon Stubbs
- Centre for Sport Science and University SportsUniversity of ViennaViennaAustria
- Department of Psychological MedicineInstitute of Psychiatry, Psychology and NeuroscienceKing's College LondonLondonUK
| | - Iain C. Campbell
- Department of Psychological MedicineCentre for Research in Eating and Weight DisordersInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| | - Ulrike Schmidt
- Department of Psychological MedicineCentre for Research in Eating and Weight DisordersInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
- Eating Disorders Outpatient UnitSouth London and Maudsley NHS Foundation TrustLondonUK
| | - Lauren Robinson
- Department of Psychological MedicineCentre for Research in Eating and Weight DisordersInstitute of Psychiatry, Psychology & NeuroscienceKing's College LondonLondonUK
| |
Collapse
|
244
|
Hackeng WM, Dreijerink KM, Brosens LA. Should we worry about high-grade pancreatic neuroendocrine tumor progression and alkylating agents? †. J Pathol 2025; 266:1-4. [PMID: 40007046 DOI: 10.1002/path.6409] [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: 12/23/2024] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
Predicting metachronous metastases in localized pancreatic neuroendocrine tumors (PanNETs) and improving survival of patients with advanced disease are some of the most important goals in PanNET research. Both are addressed by a study published recently in this journal. First, the results suggest that heterozygous DAXX mutations are already present in tumor cells but only become potentiated after a single massive chromosomal event that causes loss of heterozygosity and biallelic loss of DAXX. Second, the significant finding that the alkylating agent streptozocin may also induce a hypermutator phenotype with aggressive high-grade progression is further explored. The literature on temozolomide and peptide receptor radionuclide therapy-induced and spontaneous high-grade PanNET progression shows that the cause of high-grade progression is likely multifactorial. High-grade progressed PanNETs may show histopathological features normally seen in neuroendocrine carcinomas. Although it is not clear how often alkylating treatment induces progression, increasing evidence suggests that after an initial response, some patients indeed progress due to streptozocin or temozolomide. © 2025 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Wenzel M Hackeng
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Koen Ma Dreijerink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Lodewijk Aa Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
245
|
Navarro-Serer B, Wissler MF, Glover BK, Lerner MG, Oza HH, Wang V, Knutsdottir H, Shojaeian F, Noller K, Baskaran SG, Hughes S, Weaver AM, Wilentz D, Olayemi O, Bader JS, Fertig EJ, Gilkes DM, Wood LD. P4HA1 Mediates Hypoxia-Induced Invasion in Human Pancreatic Cancer Organoids. CANCER RESEARCH COMMUNICATIONS 2025; 5:881-895. [PMID: 40332386 PMCID: PMC12123483 DOI: 10.1158/2767-9764.crc-24-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 10/26/2024] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with dismal prognosis. PDAC develops in a hypoxic environment in which cells adapt and activate processes to allow survival under low-oxygen conditions, some of which may enhance the ability of cancer cells to invade locally or metastasize distantly. Using human PDAC organoids, we determined that hypoxia consistently enhanced invasion across 11 patient-derived models. Using RNA sequencing of hypoxic invasive organoids (compared with matched invasive normoxic organoids from the same patients), we identified prolyl 4-hydroxylase subunit alpha 1 (P4HA1) as a potential regulator of PDAC invasion in hypoxia. Leveraging publicly available datasets from human tissue, we determined that P4HA1 is more highly expressed in PDAC compared with normal pancreatic tissue and that high P4HA1 expression correlates with poor patient prognosis. To further interrogate the role of P4HA1 in invasion of hypoxic patient-derived organoids, we quantified invasion in organoids modified to knockdown or overexpress P4HA1, demonstrating that P4HA1 is necessary for hypoxia-enhanced invasion and sufficient to increase invasion in normoxia in PDAC organoids. Our results identify P4HA1 as a driver of PDAC organoid invasion in hypoxia. SIGNIFICANCE This study demonstrates that hypoxia increases invasion across a cohort of human pancreatic cancer organoids and identifies the collagen-modifying enzyme P4HA1 as a driver of hypoxia-enhanced invasion. These results characterize a molecular mechanism by which the microenvironment alters tumor cell behavior and underscore new strategies to inhibit invasion.
Collapse
Affiliation(s)
| | - Maria F. Wissler
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Brandi K. Glover
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michael G. Lerner
- Department of Physics, Engineering and Astronomy, Earlham College, Richmond, Indiana
| | - Harsh H. Oza
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Vania Wang
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Hidur Knutsdottir
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Fatemeh Shojaeian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Kathleen Noller
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Sarah Hughes
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alana M. Weaver
- Department of Physics, Engineering and Astronomy, Earlham College, Richmond, Indiana
| | - Daniel Wilentz
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | - Joel S. Bader
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
| | - Elana J. Fertig
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland
- Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, Maryland
| | - Daniele M. Gilkes
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Laura D. Wood
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
246
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025; 25:353-369. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
247
|
Trecourt A, Donzel M, Gaillot-Durand L, Bolze PA, Golfier F, Descargues P, Hajri T, Mauduit C, Devouassoux-Shisheboran M, Allias F. SALL4 as a Useful Marker for the Distinction of Various Gestational Trophoblastic Disease Subtypes: Choriocarcinoma From Other Trophoblastic Lesions and Early Complete Hydatidiform Mole From Partial Mole and NonMolar Villi. Am J Surg Pathol 2025; 49:417-428. [PMID: 39876093 PMCID: PMC11984545 DOI: 10.1097/pas.0000000000002358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
The distinction between choriocarcinoma and residual trophoblastic cell proliferation from a complete hydatidiform mole/invasive mole (CHM/IM) without villi is challenging on curettage materials. We investigated whether SALL4 immunostaining could help differentiate various gestational trophoblastic diseases. Placental site nodules (PSN; n=10), atypical PSN (APSN; n=8), placental site trophoblastic tumors (PSTT; n=9), epithelioid trophoblastic tumors (ETT; n=5), gestational choriocarcinomas (n=31), partial hydatidiform moles (PHM; n=13), CHM/IM (n=47), and nonmolar products of conception (POC) (n=26) were included. SALL4 immunostaining was quantified (0 [1% to 10%], [11% to 100%]) and characterized (scattered single-cell or clustered nuclear positivity) in 2 locations: cytotrophoblast/intermediate trophoblast and villous stromal fibroblasts. A diffuse (11% to 100%) and clustered pattern of SALL4 immunostaining in cytotrophoblast/intermediate trophoblast was statistically associated with choriocarcinomas (74.2%, 23/31) as compared with PSN (0/10; P <0.0001), APSN (0/8; P =0.0002), PSTT (0/9; P <0.0001), ETT (0/5; P =0.0034), PHM (0/13; P <0.0001), CHM/IM (0/47; P <0.0001), and nonmolar POC (0/26; P <0.0001). Most nonchoriocarcinoma samples showed no SALL4 expression; when present, it was of low level (1% to 10%) and with a scattered single-cell staining in 3/9 PSTT (33%), 1/13 PHM (7.7%), 19/47 CHM/IM (40%), and 1/26 nonmolar POC (1.7%). These results were confirmed using a validation cohort. In addition, 66% (31/47) of CHM/IM villous stromal fibroblasts showed SALL4 expression (11% to 100%) (all before 14 gestational weeks), whereas this level of expression was never observed in PHM (0/13), nor in nonmolar POC (0/26; P <0.0001). Finally, a clustered and >10% SALL4 immunostaining in cytotrophoblast/intermediate trophoblast favors choriocarcinoma diagnosis. SALL4 expression in >10% villous stromal fibroblasts before 14 gestational weeks favors CHM/IM rather than PHM and nonmolar POC.
Collapse
Affiliation(s)
- Alexis Trecourt
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre pour l’innovation en cancérologie de Lyon (CICLY), Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon-1
| | - Marie Donzel
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique (CNRS), Université Claude Bernard Lyon-1, Centre International de Recherche en Infectiologie (CIRI), UMR5308, Ecole Normale Supérieure de Lyon
| | - Lucie Gaillot-Durand
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Français de Référence des maladies trophoblastique
| | - Pierre A. Bolze
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - François Golfier
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - Pierre Descargues
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - Touria Hajri
- Centre Français de Référence des maladies trophoblastique
| | - Claire Mauduit
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale, Unité 1065, Nice, France
| | - Mojgan Devouassoux-Shisheboran
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre pour l’innovation en cancérologie de Lyon (CICLY), Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon-1
- Centre Français de Référence des maladies trophoblastique
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon
| | - Fabienne Allias
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Français de Référence des maladies trophoblastique
| |
Collapse
|
248
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [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: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
249
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
250
|
Thomas CE, Peters U. Genomic landscape of cancer in racially and ethnically diverse populations. Nat Rev Genet 2025; 26:336-349. [PMID: 39609636 DOI: 10.1038/s41576-024-00796-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2024] [Indexed: 11/30/2024]
Abstract
Cancer incidence and mortality rates can vary widely among different racial and ethnic groups, attributed to a complex interplay of genetic, environmental and social factors. Recently, substantial progress has been made in investigating hereditary genetic risk factors and in characterizing tumour genomes. However, most research has been conducted in individuals of European ancestries and, increasingly, in individuals of Asian ancestries. The study of germline and somatic genetics in cancer across racial and ethnic groups using omics technologies offers opportunities to identify similarities and differences in both heritable traits and the molecular features of cancer genomes. An improved understanding of population-specific cancer genomics, as well as translation of those findings across populations, will help reduce cancer disparities and ensure that personalized medicine and public health approaches are equitable across racial and ethnic groups.
Collapse
Affiliation(s)
- Claire E Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Epidemiology, University of Washington, Seattle, WA, USA.
| |
Collapse
|