1
|
Durmo R, Chauvie S, Minoia C, Bergesio F, Fallanca F, Peano S, Marcheselli L, Anastasia A, Boccomini C, Corradini P, Olivieri J, Arcaini L, Cavallo F, Ibatici A, Nassi L, Tarantino V, Pinto A, Stelitano C, Pulsoni A, Ricci F, Mancuso S, Cencini E, Di Renzo N, Mannarella C, Palmas A, Zinzani P, Bocci C, Rossi F, Carella AM, Federico M, Versari A, Guerra L, Luminari S. Total Metabolic Tumor Volume Is a Strong Independent Prognostic Factor in Follicular Lymphomas: Results From a Sub-Study of the FOLL12 Trial. Am J Hematol 2025. [PMID: 40366076 DOI: 10.1002/ajh.27711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 05/15/2025]
Abstract
Discordant results have been generated regarding the prognostic role of Total Metabolic Tumor Volume (TMTV) in Follicular Lymphoma (FL). The use of prospective data and the adoption of the newly defined standardized SUV4 method for calculating TMTV may generate stronger evidence. We conducted a pre-planned post hoc analysis of the prospective multicenter randomized phase III FOLL12 trial for newly diagnosed high tumor burden FL (grade 1-3a), which mandated baseline staging with PET. Baseline PET/CT scans were reviewed centrally, and TMTV was calculated using the fixed threshold of SUV4. Kaplan-Meier and Cox regression were used for survival analysis. The primary study endpoint was Progression free Survival (PFS). A total of 689 FL patients were available for TMTV definition. Median TMTV was 161 mL (IQR 50 to 388 mL) and the best cutoff value was set at 180 mL. Patients with high TMTV had a significantly lower 5-year PFS compared to those with low TMTV: 59% (95% CI, 53-65%) vs. 74% (95% CI, 69-78%) HR 1.61 (95% CI, 1.24-2.09). Prognostic role of TMTV was independent of study arm, chemotherapy regimen, and FLIPI2. Combined with FLIPI-2, we identified three groups with different 5-yr PFS rates, with the lowest rates (51%) for patients with high TMTV and high FLIPI2. Combined TMTV and FLIPI model was also prognostic to predict the risk of early progression and of death. Applying the SUV4 standard method pre-treatment TMTV is confirmed as a strong and independent predictor of PFS in FL patients. Integrating TMTV with FLIPI-2 improves risk assessment.
Collapse
Affiliation(s)
- Rexhep Durmo
- Nuclear Medicine, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | | | - Carla Minoia
- Hematology Unit, IRCCS Istituto Tumori 'Giovanni Paolo II' Bari, Bari, Italy
| | | | - Federico Fallanca
- IRCCS San Raffaele Scientific Institute, Nuclear Medicine, Milan, Italy
| | - Simona Peano
- Nuclear Medicine Division, S. Croce e Carle Hospital, Cuneo, Italy
| | | | | | - Carola Boccomini
- SC Ematologia, Dipartimento di Ematologia Ed Oncologia, AOU Città Della Salute e Della Scienza di Torino, Torino, Italy
| | | | | | - Luca Arcaini
- Department of Molecular Medicine, University of Pavia & Division of Hematology, Fondazione IRCCS Policlinico san Matteo, Pavia, Italy
| | - Federica Cavallo
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute Università di Torino, Torino, Italy
| | - Adalberto Ibatici
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Luca Nassi
- Ematologia, Az Ospedaliera Careggi, Firenze, Italy
| | | | - Antonello Pinto
- Hematology-Oncology and Stem Cell Transplantation Unit, Istituto Nazionale Tumori, IRCCS-Fondazione 'G. Pascale', Naples, Italy
| | - Caterina Stelitano
- Hematology, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | | | | | | | - Emanuele Cencini
- UOC Ematologia, Azienda Ospedaliera Universitaria Senese & University of Siena, Siena, Italy
| | | | | | - Angelo Palmas
- Ematologia e Trapianto di Midollo Osseo Ospedale 'San Francesco' Nuoro, Matera, Italy
| | - Pierluigi Zinzani
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Seràgnoli, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| | - Caterina Bocci
- U.O.S.D Ematologia Con Autotrapianto Cellule Staminali Ospedale di Civitanova Marche, Civitanova Marche, Italy
| | | | | | - Massimo Federico
- Department Chimomo, Università di Modena and Reggio Emilia, Modena, Italy
| | - Annibale Versari
- Nuclear Medicine, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Luca Guerra
- Nuclear Medicine Unit, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- University of Milano Bicocca, Milan, Italy
| | - Stefano Luminari
- Department Chimomo, Università di Modena and Reggio Emilia, Modena, Italy
- Hematology, Azienda USL IRCCS of Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
2
|
Mecklenbrauck R, Borchert N, Gabdoulline R, Poll P, Funke C, Brandes M, Dallmann LK, Fiedler W, Krauter J, Trummer A, Hertenstein B, Müller M, Lübbert M, Schwalenberg M, Voss A, Di Donato N, Bergmann A, Gaidzik V, Döhner K, Döhner H, Ganser A, Heidel FH, Thol FR, Heuser M. Prognostic impact of clonal representation of myelodysplasia-related gene mutations in acute myeloid leukemia. Leukemia 2025:10.1038/s41375-025-02622-6. [PMID: 40295828 DOI: 10.1038/s41375-025-02622-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Affiliation(s)
- Rabea Mecklenbrauck
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nora Borchert
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Razif Gabdoulline
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Piroska Poll
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Carolin Funke
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Maximilian Brandes
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Louisa-Kristin Dallmann
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Walter Fiedler
- Department of Internal Medicine, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Jürgen Krauter
- Department of Hematology and Oncology, Municipal Hospital Braunschweig, Braunschweig, Germany
| | - Arne Trummer
- Department of Hematology, Oncology and Palliative Care, Heidekreisklinikum, Walsrode, Germany
| | - Bernd Hertenstein
- Department of Internal Medicine I, Hospital Bremen-Mitte, Bremen, Germany
| | - Martin Müller
- Department of Hematology, Oncology and Immunology, Hospital Siloah, Hannover, Germany
| | - Michael Lübbert
- Department of Internal Medicine I, University Hospital Freiburg, Freiburg, Germany
| | - Monika Schwalenberg
- Department of Hematology, Oncology and Palliative Care, Hospital Lüdenscheid, Lüdenscheid, Germany
| | - Andreas Voss
- Department of Oncology and Hematology, Klinikum Oldenburg, Oldenburg, Germany
| | | | - Anke Bergmann
- Institute of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Verena Gaidzik
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Arnold Ganser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Florian H Heidel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
- Leibniz Institute on Aging, Fritz-Lipmann Institute, Jena, Germany
| | - Felicitas R Thol
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.
- Department of Internal Medicine IV, University Hospital Halle (Saale), Martin-Luther-University Halle-Wittenberg, Halle, Germany.
| |
Collapse
|
3
|
Li X, Pan L, Li W, Liu B, Xiao C, Chew V, Zhang X, Long W, Ginhoux F, Loscalzo J, Buggert M, Zhang X, Sheng R, Wang Z. Deciphering immune predictors of immunotherapy response: A multiomics approach at the pan-cancer level. Cell Rep Med 2025; 6:101992. [PMID: 40054456 PMCID: PMC12047473 DOI: 10.1016/j.xcrm.2025.101992] [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: 08/03/2024] [Revised: 01/15/2025] [Accepted: 02/05/2025] [Indexed: 04/18/2025]
Abstract
Immune checkpoint blockade (ICB) therapy has transformed cancer treatment, yet many patients fail to respond. Employing single-cell multiomics, we unveil T cell dynamics influencing ICB response across 480 pan-cancer and 27 normal tissue samples. We identify four immunotherapy response-associated T cells (IRATs) linked to responsiveness or resistance and analyze their pseudotemporal patterns, regulatory mechanisms, and T cell receptor clonal expansion profiles specific to each response. Notably, transforming growth factor β1 (TGF-β1)+ CD4+ and Temra CD8+ T cells negatively correlate with therapy response, in stark contrast to the positive response associated with CXCL13+ CD4+ and CD8+ T cells. Validation with a cohort of 23 colorectal cancer (CRC) samples confirms the significant impact of TGF-β1+ CD4+ and CXCL13+ CD4+ and CD8+ T cells on ICB efficacy. Our study highlights the effectiveness of single-cell multiomics in pinpointing immune markers predictive of immunotherapy outcomes, providing an important resource for crafting targeted immunotherapies for successful ICB treatment across cancers.
Collapse
Affiliation(s)
- Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, China; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning 110122, China; Department of Physiology and Pharmacology, Karolinska Institutet, 171 65 Solna, Sweden.
| | - Lu Pan
- Institute of Environmental Medicine, Karolinska Institutet, 171 65 Solna, Sweden
| | - Weiyuan Li
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, China; Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650021, China
| | - Bingyang Liu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming, Yunnan 650091, China
| | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth-Duke NUS Academic Medical Centre, Singapore 169856, Singapore
| | - Xuan Zhang
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Wang Long
- Department of Pathology, Nihon University, Tokyo 102-0074, Japan
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore 138648, Singapore; Institut Gustave Roussy, INSERM U1015, Bâtiment de Médecine Moléculaire 114 rue Edouard Vaillant, 94800 Villejuif, France; Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, 141 52 Huddinge, Sweden
| | - Xiaolu Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong 518057, China.
| | - Ren Sheng
- College of Life and Health Sciences, Northeastern University, Shenyang, Liaoning 110819, China; School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510000, China.
| | - Zhenning Wang
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning 110122, China; The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
4
|
Zalfa F, Manca P, Carotti S, Vallese S, Righi D, Taffon C, Nibid L, Sbaraglia M, Rabitti C, Pantano F, Tonini G, Dei Tos AP, Vincenzi B, Perrone G. A Nanostring gene expression approach identifies aggressive clinical behavior related genes in dedifferentiated liposarcoma. Sci Rep 2025; 15:9204. [PMID: 40097500 PMCID: PMC11914264 DOI: 10.1038/s41598-025-91791-6] [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: 09/22/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Dedifferentiated liposarcoma (DDLPS) is one of the most common subtypes of soft tissue sarcoma with a highly variable clinical behavior. Despite advanced molecular approaches are exploring the genetic panorama of DDLPS progression, to date the driver genes of the aggressive clinical behavior in DDLPS have not been identified yet. Here, we used a Nanostring nCounter approach to study the gene expression profile of 60 selected genes involved in DDLPS progression, in a cohort of DDLPS with aggressive clinical behavior, in comparison to a cohort of DDLPS with indolent clinical behavior. We identified five genes whose expression is significantly and consistently altered in aggressive compared to indolent DDLPS. Moreover, by a clinical outcome analyses we found MAP3K12 gene expression linked with both a higher risk of metastases and death. We envisage that the identified genes could represent the first genes of a genetic signature able to predict the clinical evolution of a DDLPS.
Collapse
Affiliation(s)
- Francesca Zalfa
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy.
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy.
| | - Paolo Manca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Simone Carotti
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Silvia Vallese
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Daniela Righi
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Chiara Taffon
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| | - Lorenzo Nibid
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| | - Marta Sbaraglia
- Department of Medicine DIMED, University of Padua, Padua, Italy
- Pathology Unit, University-Hospital of Padua, Padua, Italy
| | - Carla Rabitti
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesco Pantano
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Giuseppe Tonini
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine DIMED, University of Padua, Padua, Italy
- Pathology Unit, University-Hospital of Padua, Padua, Italy
| | - Bruno Vincenzi
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| |
Collapse
|
5
|
Chung S, Kim C. Comparative Analysis of Transcription Factors TWIST2, GATA3, and HES5 in Glioblastoma Multiforme : Evaluating Biomarker Potential and Therapeutic Targets Using in Silico Methods. J Korean Neurosurg Soc 2025; 68:202-212. [PMID: 39444320 PMCID: PMC11924635 DOI: 10.3340/jkns.2024.0149] [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: 08/07/2024] [Accepted: 10/23/2024] [Indexed: 10/25/2024] Open
Abstract
OBJECTIVE Glioblastoma multiforme (GBM) is characterized by substantial heterogeneity and limited therapeutic options. As molecular approaches to central nervous system tumors have gained prominence, this study examined the roles of three genes, TWIST2, GATA3, and HES5, known to be involved in oncogenesis, developmental processes, and maintenance of cancer stem cell properties, which have not yet been extensively studied in GBM. This study is the first to present gene expression data for TWIST2, GATA3, and HES5 specifically within the context of GBM patient survival. METHODS Gene expression data for TWIST2, GATA3, and HES5 were collected from GBM and normal brain tissues using datasets from The Cancer Genome Atlas via the Genomic Data Commons portal and the Genotype-Tissue Expression database. These data were rigorously analyzed using in silico methods. RESULTS All three genes were significantly more expressed in GBM tissues than in normal tissues. TWIST2 and GATA3 were linked to lower survival rates in GBM patients. Interestingly, higher HES5 levels were associated with better survival rates, suggesting a complex role that needs more investigation. CONCLUSION This study shows that TWIST2, GATA3, and HES5 could help predict outcomes in GBM patients. Our multigene model offers a better understanding of GBM and points to new treatment options, bringing hope for improved therapies and patient outcomes. This research advances our knowledge of GBM and highlights the potential of molecular diagnostics in oncology.
Collapse
Affiliation(s)
- Suhmi Chung
- Department of Neurosurgery, Kangwon National University Hospital, Chuncheon, Korea
| | - Choonghyo Kim
- Department of Neurosurgery, Kangwon National University Hospital, Chuncheon, Korea
- College of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
6
|
Li J, Li Z, Wang Y, Li Y, Zhang J, Li Z, Tang L. CT radiomics-based intratumoral and intertumoral heterogeneity indicators for prognosis prediction in gastric cancer patients receiving neoadjuvant chemotherapy. Eur Radiol 2025:10.1007/s00330-025-11430-6. [PMID: 39953151 DOI: 10.1007/s00330-025-11430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/30/2024] [Accepted: 01/13/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVES CT-based intratumoral and intertumoral heterogeneity indicators were integrated to develop a prognostic model for locally advanced gastric cancer (LAGC) patients undergoing neoadjuvant chemotherapy (NACT). METHODS This retrospective study included 568 LAGC patients treated with NACT from two hospitals. The intratumor heterogeneity score (ITHscore) was developed to quantify the intratumoral heterogeneity of LAGCs on CT; intertumoral heterogeneity was characterized by combining the primary tumor (PT) and lymph node (LN) sizes on CT. CT indicators were measured on baseline and posttreatment CT scans; the reduction rates (%Δ) were calculated. The overall survival (OS) of all patients was recorded. Cox regression analysis was used to construct a preoperative survival prediction model (Pre-SPM) based on the baseline indicators and %Δ indicators. The predictive performance of Pre-SPM for OS was assessed. The clinicopathological data, including the ypTNM stage, were also collected to evaluate their impact on OS. RESULTS Patients with lower baseline ITHscore had better prognoses (p < 0.001). Approximately 13.01% of patients exhibited contradictory changes in PT and LN sizes. Cox regression analysis selected the baseline ITHscore, baseline PT area, %ΔPT, and %ΔLN to establish the Pre-SPM. In the external validation cohort, the c-index of Pre-SPM for predicting OS was 0.72, while the AUC for predicting 5-year OS was 0.73. After adjusting for the influence of clinicopathological features, including the ypTNM stage, Pre-SPM remained an independent prognostic factor. CONCLUSION The Pre-SPM model, combining intratumoral heterogeneity and intertumoral heterogeneity, has the potential to predict the OS of LAGC patients receiving NACT. KEY POINTS Question Increased tumor heterogeneity in LAGC affects prognosis, but effective non-invasive CT methods for assessing intratumoral and intertumoral heterogeneity are lacking. Findings ITHscore indicates intratumoral heterogeneity, while changes in PT and LN sizes reflect intertumoral heterogeneity. The Pre-SPM model, integrating both, independently predicts patient outcomes. Clinical relevance Pre-SPM enhances prognosis prediction by quantifying intratumoral and intertumoral heterogeneity, potentially guiding more personalized and effective treatment strategies for patients with LAGC.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yinkui Wang
- Department of Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuzhuo Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Jing Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing, China.
| | - Ziyu Li
- Department of Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
7
|
Balcar L, Dominik N, Mozayani B, Semmler G, Halilbasic E, Mandorfer M, Reiberger T, Trauner M, Scheiner B, Stättermayer AF. Elevated Hepatic Copper Content in Porto-Sinusoidal Vascular Disorder (PSVD): Leading Down a Wrong Track. Liver Int 2025; 45:e16175. [PMID: 39807082 PMCID: PMC11730389 DOI: 10.1111/liv.16175] [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: 08/14/2024] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND AND AIMS Porto-sinusoidal vascular disorder (PSVD) is a rare vascular liver disorder characterised by specific histological findings in the absence of cirrhosis, which is poorly understood in terms of pathophysiology. While elevated hepatic copper content serves as diagnostic hallmark in Wilson disease (WD), hepatic copper content has not yet been investigated in PSVD. METHODS Patients with a verified diagnosis of PSVD at the Medical University of Vienna and available hepatic copper content at the time of diagnosis of PSVD were retrospectively included. Elevated hepatic copper content was correlated with cholestatic changes and WD diagnostics in PSVD and analysed for liver-related outcomes (first/further hepatic decompensation/liver-related death). RESULTS Overall, 92 patients were included into this study (mean age 49 ± 16; 57% male; median hepatic copper content was 30 [IQR: 18-55] μg/g) of whom 29 (32%) had moderately (≥ 50 μg/g) and 4 (4%) strongly (≥ 250 μg/g) elevated hepatic copper content. Elevated levels of hepatic copper were associated with younger age in multivariable linear regression analysis. After adjusting for age, decompensation status and albumin, hepatic copper content was significantly associated with the outcome of interest (log, per 10; aHR: 1.60 [95% CI: 1.14-2.25]; p = 0.007). A hepatic copper cut-off at ≥ 90 μg/g identified PSVD patients with considerable risk of liver-related outcomes (at 2 years: 51% vs. 12%). CONCLUSION Elevated hepatic copper seems frequent in patients with PSVD even in the absence of cholestatic features, especially in young patients, which makes differential diagnosis to WD challenging. Since PSVD patients with elevated hepatic copper content had increased risk for liver-related outcomes, the pathomechanisms underlying hepatic copper accumulation in PSVD should be investigated as this may open new therapeutic avenues.
Collapse
Affiliation(s)
- Lorenz Balcar
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Nina Dominik
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Behrang Mozayani
- Department of PathologyMedical University of ViennaViennaAustria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Emina Halilbasic
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Bernhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Clinical Research Group MOTIONMedical University of ViennaViennaAustria
| | - Albert Friedrich Stättermayer
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| |
Collapse
|
8
|
Galtier J, Mesguich C, Sesques P, Dupont V, Bachy E, Di Blasi R, Thieblemont C, Gastinne T, Cartron G, Brisou G, Gros F, Decroocq J, Morschhauser F, Rubio M, Drieu La Rochelle L, Le Bras F, Carras S, Chauchet A, Bay J, Joris M, Loschi M, Tanguy‐Schmidt A, Marquet A, Camus V, Le Gouill S, Houot R, Bouabdallah K. Outcomes of patients with relapsed or refractory primary mediastinal B-cell lymphoma treated with anti-CD19 CAR-T cells: CARTHYM, a study from the French national DESCAR-T registry. Hemasphere 2025; 9:e70091. [PMID: 39968186 PMCID: PMC11833168 DOI: 10.1002/hem3.70091] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/03/2025] [Accepted: 01/08/2025] [Indexed: 02/20/2025] Open
Abstract
Primary mediastinal B-cell lymphoma (PMBL) is often cured with dose-dense anthracycline-based regimens but the prognosis at relapse or progression remains poor. While anti-CD19 CAR-T cell therapy has dramatically improved outcomes in relapsed or refractory large B-cell lymphoma, far less is known about their efficacy in PMBL. Using the systematic record of all patients treated with CAR-T cells prospectively included in the DESCAR-T registry in France, along with centrally reviewed positon-emission tomography (PET) imaging, we describe the outcomes and key determinants of treatment success in PMBL patients treated over a 6-year period. Among 82 patients infused in the registry we observed a best complete response (CR) rate, 2-year progression-free survival (PFS), and 2-year overall survival (OS) of 68.1%, 57.4%, and 73.8%, respectively. Outcomes were even better for the 62 patients infused with axicabtagene ciloleucel, with best CR rate, 2-year PFS, and 2-year OS reaching 74.5%, 70.4%, and 86.9%, respectively. Achieving a Deauville score of 1-4 or a ΔSUVmax reduction of more than 24% at the 1-month evaluation was associated with excellent outcomes, whereas increased total metabolic tumor volume baseline PET increased the risk of treatment failure. Surprisingly, neither the response to bridging therapy nor the type of bridging therapy (chemotherapy versus immune checkpoint inhibitors) were associated with long-term outcomes. In conclusion, this study confirms that anti-CD19 CAR-T cells as a valid standard-of-care for relapsed and refractory PMBL and highlights key determinants of treatment success.
Collapse
Affiliation(s)
- Jean Galtier
- CHU de Bordeaux, Service d'hématologie Clinique et de thérapie cellulaireBordeauxFrance
| | | | - Pierre Sesques
- Hospices civils de Lyon, Service d'hématologie cliniqueLyonFrance
| | | | - Emmanuel Bachy
- Hospices civils de Lyon, Service d'hématologie cliniqueLyonFrance
| | - Roberta Di Blasi
- Hôpital Saint‐Louis, Service d'hématologie clinique, Assistance publique ‐ Hôpitaux de ParisParisFrance
| | - Catherine Thieblemont
- Hôpital Saint‐Louis, Service d'hématologie clinique, Assistance publique ‐ Hôpitaux de ParisParisFrance
| | | | | | - Gabriel Brisou
- Institut Paoli Calmettes, Service d'hématologie cliniqueMarseilleFrance
| | - François‐Xavier Gros
- CHU de Bordeaux, Service d'hématologie Clinique et de thérapie cellulaireBordeauxFrance
| | - Justine Decroocq
- Hôpital Cochin, Service d'hématologie clinique, Asistance publique ‐ Hôpitaux de ParisParisFrance
| | | | | | | | - Fabien Le Bras
- Hôpital Henri Mondor, Service d'hématologie clinique, Assistance publique ‐ Hôpitaux de ParisParisFrance
| | - Sylvain Carras
- CHU de Grenoble, Service d'hématologie cliniqueGrenobleFrance
| | - Adrien Chauchet
- CHU de Besançon, Service d'hématologie cliniqueBensaçonFrance
| | - Jacques‐Olivier Bay
- CHU de Clermont‐Ferrand, Service d'hématologie cliniqueClermont‐FerrandFrance
| | - Magalie Joris
- CHU d'Amiens, Service d'hématologie cliniqueAmiensFrance
| | | | | | | | - Vincent Camus
- Département d'HématologieCentre Henri BecquerelRouenFrance
| | | | - Roch Houot
- CHU de Rennes, Service d'hématologie cliniqueRennesFrance
| | - Krimo Bouabdallah
- CHU de Bordeaux, Service d'hématologie Clinique et de thérapie cellulaireBordeauxFrance
| |
Collapse
|
9
|
Wang Q, Zhong W, Xiao Y, Lin G, Lu J, Xu L, Zhang G, Liu A, Du J, Wu B. Pan-immune-inflammation value predicts immunotherapy response and reflects local antitumor immune response in rectal cancer. Cancer Sci 2025; 116:350-366. [PMID: 39601159 PMCID: PMC11786305 DOI: 10.1111/cas.16400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
The pan-immune-inflammation value reflects the systemic inflammatory response, and tumor-infiltrating lymphocytes indicate a local immune response in rectal cancer. However, the association between systemic inflammatory response, as indicated by the pan-immune-inflammation value, and local immune responses in rectal cancer remains unclear. This study analyzed 915 treatment-naïve rectal cancer patients from the Peking Union Medical College Hospital and PLA General Hospital (PLAGH) cohorts who underwent radical surgery to investigate the relationship between the pan-immune-inflammation value and immune responses. Lower pan-immune-inflammation value was significantly associated with improved disease-free survival and cancer-specific survival. Multivariate Cox regression models identified the pan-immune-inflammation value as an independent prognostic factor. In the PLAGH cohort, patients with low pan-immune-inflammation values had higher immune cell levels, activated immune pathways, and increased expression of immune checkpoint genes according to RNA sequencing. Hematoxylin and eosin staining and immunohistochemical analysis revealed that lower pan-immune-inflammation value was associated with higher tumor-infiltrating lymphocyte density, more mature tertiary lymphoid structures, increased CD8+ T cells, and elevated human lymphocyte antigen class I expression. Conversely, patients with high pan-immune-inflammation values exhibited pathways linked to tumor progression, such as angiogenesis, epithelial-mesenchymal transition, hypoxia, KRAS signaling, and TGF-ß signaling. Among patients receiving anti-PD-1 therapy, responders had low pre- and post-treatment pan-immune-inflammation values. The pan-immune-inflammation value is a reliable marker associated with distinct immune microenvironment characteristics and can effectively predict disease-free survival, cancer-specific survival, and response to immunotherapy.
Collapse
Affiliation(s)
- Qianyu Wang
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Wentao Zhong
- Medical Department of General Surgery, The First Medical CenterChinese PLA General HospitalBeijingChina
| | - Yi Xiao
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Guole Lin
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junyang Lu
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lai Xu
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Guannan Zhang
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Aijun Liu
- Department of Pathology, The 7th Medical CenterChinese PLA General HospitalBeijingChina
| | - Junfeng Du
- Medical Department of General Surgery, The First Medical CenterChinese PLA General HospitalBeijingChina
| | - Bin Wu
- Department of General Surgery, Peking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
10
|
Qu Y, Shourabizadeh H, Subramanian A, Aleman DM, Rousseau LM, Law AD, Viswabandya A, Michelis FV. Differential impact of CD34+ cell dose for different age groups in allogeneic hematopoietic cell transplantation for acute leukemia: a machine learning-based discovery. Exp Hematol 2025; 141:104684. [PMID: 39581301 DOI: 10.1016/j.exphem.2024.104684] [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: 05/20/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024]
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) presents a potentially curative treatment for hematologic malignancies yet carries associated risks and complications. Continuous research focuses on predicting outcomes and identifying risk factors. Notably, the influence of CD34+ cell dose on overall survival (OS) has been the subject of numerous studies yielding contradictory results. We developed machine learning (ML) models to predict allo-HCT outcomes and, through the application of SHapley Additive exPlanations (SHAP), an explainable artificial intelligence (XAI) technique enabled the identification of new and clinically relevant feature-outcome relationships. In particular, we identified a clear interaction between CD34+ cell dose of peripheral blood stem cell (PBSC) grafts and patient age at allo-HCT for patients with acute leukemia. Results of multivariable analysis validated the interaction effect: in young patients with acute leukemia (aged ≤45 years), low dose of CD34+ cells (<4.3 × 106 CD34+/kg) was associated with better OS against high dose (≥7 ×106 CD34+/kg) (hazard ratio [HR], 0.38; p = 0.019), while for older patients with acute leukemia (>45 years), low CD34+ cell dose (<3.8 ×106 CD34+/kg) was associated with worse OS against high dose (≥6.1 ×106 CD34+/kg) (HR, 1.58; p = 0.033). In conclusion, our findings suggest that tailoring CD34+ cell dose by patient age may benefit patients with acute leukemia undergoing allo-HCT, while XAI showcases excellent proficiency in revealing such interactions.
Collapse
Affiliation(s)
- Yiyang Qu
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Hamed Shourabizadeh
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Aravind Subramanian
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Dionne M Aleman
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Louis-Martin Rousseau
- Department of Mathematical and Industrial Engineering, Polytechnique Montreal, Montreal, Quebec, Canada
| | - Arjun D Law
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Auro Viswabandya
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Fotios V Michelis
- Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
da Costa Pereira JP, Prado CM, Gonzalez MC, Cabral PC, de Oliveira Guedes FF, da Silva Diniz A, Fayh APT. Prognostic significance of novel muscle quality index utilization in hospitalized adults with cancer: A secondary analysis. JPEN J Parenter Enteral Nutr 2025; 49:112-121. [PMID: 39503074 DOI: 10.1002/jpen.2701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND This study aimed to investigate and propose novel approaches to calculate muscle quality index (MQI) using muscle mass derived from single-frequency bioelectrical impedance analysis (SF-BIA) and calf circumference in both unadjusted and body mass index (BMI)-adjusted forms. In addition, we examined their prognostic significance in patients with cancer. METHODS A secondary analysis was conducted on a prospective cohort study of patients with cancer. Handgrip strength was measured. SF-BIA was conducted to estimate appendicular lean soft tissue (ALST, in kilograms). MQI was calculated using three approaches: (1) the ratio of handgrip strength to ALST (MQISF-BIA), (2) the ratio of handgrip strength to calf circumference (MQIcalf circumference), and (3) the ratio of handgrip strength to BMI-adjusted calf circumference (MQIadj. calf circumference). Maximally selected log-rank was calculated to estimate their cutoff values to predict survival. RESULTS Two hundred eighty-four patients were included (51.1% men; median age, 61 years). Solid tumors were the most frequent (89.8%). All approaches to MQI (MQISF-BIA, MQIcalf circumference, and MQIadj. calf circumference) were independent predictors of 6-month mortality. The found cutoffs were (1) MQISF-BIA (<1.52 for men, <0.63 for women), (2) MQIcalf circumference (<0.74 for men, <0.24 for women), and (3) MQIadj. calf circumference (<0.75 for men, <0.25 for women). CONCLUSION This study introduces MQISF-BIA, MQIcalf circumference, and MQIadj. calf circumference as future potential surrogate methods for computing MQI in clinical practice when other robust procedures are unavailable, pending further validation.
Collapse
Affiliation(s)
- Jarson P da Costa Pereira
- Department of Nutrition, Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Carla M Prado
- Department of Agricultural, Food and Nutritional Science, Human Nutrition Research Unit, University of Alberta, Edmonton, Alberta, Canada
| | - M Cristina Gonzalez
- Postgraduate Program in Nutrition and Food, Federal University of Pelotas, Pelotas, Brazil
| | - Poliana C Cabral
- Department of Nutrition, Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Brazil
| | | | - Alcides da Silva Diniz
- Department of Nutrition, Postgraduate Program in Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Ana P T Fayh
- Postgraduate Program in Health Science, Health Science Center, Federal University of Rio Grande do Norte, Natal, Brazil
- PesqClin Lab, Onofre Lopes University Hospital, Brazilian Company of Hospital Services (EBSERH), Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
12
|
Midik MM, Gunenc D, Acar PF, Karaca BS. Prognostic Value of Blood-Based Inflammatory Markers in Cancer Patients Receiving Immune Checkpoint Inhibitors. Cancers (Basel) 2024; 17:37. [PMID: 39796668 PMCID: PMC11719015 DOI: 10.3390/cancers17010037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Although immune checkpoint inhibitors (ICIs) have significantly improved cancer treatment, a substantial proportion of patients do not benefit from these therapies, revealing the crucial need to identify reliable biomarkers. Inflammatory markers, such as the neutrophil-to-lymphocyte ratio (NLR), systemic immune-inflammation index (SII), pan-immune inflammation value (PIV), systemic inflammation response index (SIRI), lactate dehydrogenase (LDH), and C-reactive protein (CRP), may provide insights into treatment outcomes. Objectives: This study aimed to evaluate the prognostic value of multiple inflammatory markers in patients with cancer receiving ICI-based therapies. Methods: A retrospective analysis was performed on 226 patients treated with ICI-based therapies at a single center between 2012 and 2023. The inflammatory markers NLR, PIV, SII, SIRI, LDH, CRP, and albumin were assessed. Cut-off values were determined using maximally selected rank statistics, and overall survival (OS) and progression-free survival (PFS) were evaluated using the Kaplan-Meier method and Cox regression analysis. Results: High NLR, PIV, SII, SIRI, LDH, and CRP, as well as low albumin levels, were associated with worse OS and PFS (p < 0.001). In the multivariate analysis, high CRP, LDH, NLR, PIV, and SII independently predicted worse OS. Conclusions: Our findings confirm the prognostic utility of several inflammatory biomarkers in patients with cancer receiving ICIs, highlighting their potential for treatment stratification. Further studies are necessary to standardize cut-off values and validate these findings across broader, more diverse populations.
Collapse
Affiliation(s)
| | | | | | - Burcak Saziye Karaca
- Division of Medical Oncology, Department of Internal Medicine, Medical Faculty, Ege University, 35100 Izmir, Turkey; (M.M.M.); (D.G.); (P.F.A.)
| |
Collapse
|
13
|
Zhao A, Tu D, He Y, Liu L, Wu B, Ren Y. Identification and validation the predictive biomarkers based on risk-adjusted control chart in gemcitabine with or without erlotinib for pancreatic cancer therapy. Front Genet 2024; 15:1497254. [PMID: 39741907 PMCID: PMC11685217 DOI: 10.3389/fgene.2024.1497254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
Background In a randomized clinical controlled trial (PA.3) conducted by the Canadian Cancer Trials Group, the effects of gemcitabine combined with the targeted drug erlotinib (GEM-E) versus gemcitabine alone (GEM) on patients with unresectable, locally advanced, or metastatic pancreatic cancer were studied. This trial statistically demonstrated that the GEM-E combination therapy moderately improves overall survival (OS) of patients. However, real-world analysis suggested that GEM-E for pancreatic cancer was not more effective than GEM. The heterogeneity in outcomes or treatment effect exist. Thus, we tried to find predictive biomarkers to identifying the heterogeneous patients. Methods Of the 569 eligible patients, 480 patients had plasma samples. Univariate and multivariate Cox proportional hazards model were used to identify baseline characteristics related to OS, and a risk adjusted Exponentially Weighted Moving Average (EWMA) control chart based on a weighted score test from the Cox model was constructed to monitor patients' survival risk. Maximally selected rank statistics were constructed to identifying the predictive biomarkers, in addition, a risk adjusted control chart based on a weighted score test from the Cox model was constructed to validating the predictive biomarkers, discover the patients who sensitive to the GEM-E or GEM. Results Three baseline characteristics (ECOG performance status, extent of disease, and pain intensity) were identified related to prognosis. A risk-adjusted EWMA control chart was constructed and showed that GEM-E did improve OS in a few patients. Three biomarkers (BMP2, CXCL6, and HER2) were identified as predictive biomarkers based on maximum selected rank test, and using the risk-adjusted EWMA control chart to validate the reality and discover some patients who are sensitive to the GEM-E therapy. Conclusion In reality, GEM-E has not shown a significant advantage over GEM in the treatment of pancreatic cancer. However, we discovered some patients who are sensitive to the GEM-E therapy based on the predictive biomarkers, which suggest that the predictive biomarkers provide ideas for personalized medicine in pancreatic cancer.
Collapse
Affiliation(s)
- Aijun Zhao
- School of Mathematical Science and Geomathematics Key Laboratory of Sichuan Province, Chengdu University of Technology, Chengdu, China
| | - Dongsheng Tu
- Department of Public Health Sciences, Canadian Cancer Trials Group, Queen’s University, Kingston, ON, Canada
| | - Ye He
- Visual Computing and Virtual Reality Key Laboratory of Sichuan Province, Sichuan Normal University, Chengdu, China
| | - Liu Liu
- School of Mathematical Science and Geomathematics Key Laboratory of Sichuan Province, Chengdu University of Technology, Chengdu, China
| | - Bin Wu
- College of Management Science, Chengdu University of Technology, Chengdu, China
| | - Yixing Ren
- Department of General Surgery, Institute of Hepato-Biliary-Pancreas and Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
14
|
Zhang R, Datta S. asmbPLS: biomarker identification and patient survival prediction with multi-omics data. Front Genet 2024; 15:1444054. [PMID: 39649094 PMCID: PMC11621212 DOI: 10.3389/fgene.2024.1444054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024] Open
Abstract
Introduction With the advancement of high-throughput studies, an increasing wealth of high-dimensional multi-omics data is being collected from the same patient cohort. However, leveraging this multi-omics data to predict survival outcomes poses a significant challenge due to its complex structure. Methods In this article, we present a novel approach, the Adaptive Sparse Multi-Block Partial Least Squares (asmbPLS) Regression model, which introduces a dynamic assignment of penalty factors to distinct blocks within various PLS components, facilitating effective feature selection and prediction. Results We compared the proposed method with several state-of-the-art algorithms encompassing prediction performance, feature selection and computation efficiency. We conducted comprehensive evaluations using both simulated data with various scenarios and a real dataset from the melanoma patients to validate the effectiveness and efficiency of the asmbPLS method. Additionally, we applied the lung squamous cell carcinoma (LUSC) dataset from The Cancer Genome Atlas (TCGA) to further assess the feature selection capability of asmbPLS. Discussion The inherent nature of asmbPLS imparts it with higher sensitivity in feature selection compared to other methods. Furthermore, an R package called asmbPLS implementing this method is made publicly available.
Collapse
Affiliation(s)
| | - Susmita Datta
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
15
|
Tang SY, Lin C, Ma HP, Chen TY, Lo MT, Kuo PH, Hsu HH, Wu CK, Peng CK, Lin YT, Tsai CH, Lin YH. Implication of heart rhythm complexity in predicting long-term outcomes in pulmonary hypertension. J Formos Med Assoc 2024:S0929-6646(24)00516-3. [PMID: 39510914 DOI: 10.1016/j.jfma.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/04/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Pulmonary hypertension (PH) is a serious disease, however simple tools to predict outcomes are lacking. In our previous investigation, we found that heart rate variability (HRV) and heart rhythm complexity (HRC) were associated with the detection and severity of PH, however their association with PH mortality remains unclear. The aim of this study was to investigate these metrics as a tool for determining long-term outcomes in PH patients. We enrolled 74 Asian PH patients with WHO PH group 1 or 4 at a single hospital in Taiwan between March 2012 and June 2018. After a median follow-up duration of 58 months (to January 2023), 22 patients had died. The patients who died had a significantly lower lean body mass index (BMI), impaired renal function, higher N-terminal pro B-type natriuretic peptide (NT-proBNP) level, lower very low-frequency (VLF), lower short-term detrended fluctuation analysis α1 (DFAα1), and lower multiscale entropy scale 5 value. In multivariable analysis, BMI, VLF and multiscale entropy scale 5 were significantly associated with survival. The best cut-off VLF and scale 5 values were 115.13 and 0.738, respectively. We then categorized the study population into three groups: both elevated VLF/scale 5 (group 1), either depressed VLF or depressed scale 5 (group 2), and both depressed VLF/scale 5 (group 3). The results showed that group 1 had the best outcomes, whereas group 3 had the worst survival (P < 0.001). Combining HRV and HRC metrics appears to be a good non-invasive tool to predict the long-term outcomes of patients with PH.
Collapse
Affiliation(s)
- Shu-Yu Tang
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Chen Lin
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Hsi-Pin Ma
- Department of Electrical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Tsung-Yan Chen
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu, Taiwan
| | - Men-Tzung Lo
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Ping-Hung Kuo
- Division of Pulmonology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsao-Hsun Hsu
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan; National Taiwan University Cancer Center, Taipei, 106, Taiwan
| | - Cho-Kai Wu
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Kang Peng
- Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston MA, 02115, USA
| | - Yen-Tin Lin
- Division of Cardiology, Department of Internal Medicine, Taoyuan General Hospital, Taoyuan, Taiwan.
| | - Cheng-Hsuan Tsai
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Yen-Hung Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
16
|
Zhang T, Gao C, Liu J, Fu G, Li B, Liu H, Zhang R, Wang P, Ning Z, Yang B, Chu H, He B, Zhang J, Zhou L, Li Y, Zhang Y, Hu H, Xu Y, Zeng J, Guo J, Su X, Soliman O, Serruys PW, Tao L. Impact of Operator Experience on Left Atrial Appendage Occlusion Outcomes. JACC Clin Electrophysiol 2024; 10:2461-2470. [PMID: 39269400 DOI: 10.1016/j.jacep.2024.07.010] [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: 05/06/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The relationship between long-term outcomes and operator experience for left atrial appendage occlusion (LAAO) is still unknown. OBJECTIVES This study sought to explore the association between operator LAAO experience and one-year clinical outcomes. METHODS The RECORD study (Registry to Evaluate Chinese Real-World Clinical Outcomes in Patients With AF Using the WATCHMAN Left Atrial Appendage Closure Technology; NCT03917563) was a multicenter, prospective registry that included patients with the WATCHMAN LAAO device (Boston Scientific) in China from April 1, 2019, to October 31, 2020. The current analyses included patients with solely LAAO from the registry; those who had concomitant LAAO and ablation/other procedures were excluded. The primary outcome was a composite endpoint of death, stroke, systemic embolism, and Bleeding Academic Research Consortium (BARC)-defined type 3 or 5 bleeding at 1 year. RESULTS A total of 1,547 LAAO patients and 111 operators were included. The mean CHA2DS2-VASc and HAS-BLED scores of patients were 4.0 ± 1.8 and 2.5 ± 1.1, respectively. The mean age of operators was 47.0 ± 7.2 years, 15 (13.5%) were female, and 52 (46.8%) were electrophysiologists. Utilizing maximally selected log-rank statistics, the thresholds to categorize an experienced operator were performing ≥32 LAAOs annually or ≥134 LAAOs in total. Performing ≥32 LAAOs annually is the better criterion than ≥134 LAAOs in total (absolute net reclassification index: 25.79%; P < 0.001). Compared with the ≥32 LAAO annually group, the <32 group was associated with a 1.8-fold (HRadjusted: 1.79; 95% CI: 1.16-2.78; P = 0.009) increase in the risk of the primary endpoint, and such risk in the <32 group can be reduced by ∼12% after performing each additional 5 cases (HRadjusted per 5 cases: 0.88; 95% CI: 0.78-0.99; P = 0.033). CONCLUSIONS Performing ≥32 LAAOs annually could be a threshold to categorize an experienced operator. Before reaching this threshold, the risk of death, stroke, systemic embolism, and BARC-defined type 3 or 5 bleeding decreased by 12% after every 5 cases performed.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chao Gao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Jianzheng Liu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guotao Fu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Boyu Li
- Biomedical Engineering, University of Shanghai For Science and Technology, Shanghai, China
| | - Haitao Liu
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ruining Zhang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ping Wang
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhongping Ning
- Department of Cardiology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Bing Yang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huimin Chu
- Department of Cardiology, Arrhythmia Center, Ningbo First Hospital, Ningbo, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ling Zhou
- Department of Cardiology, Nanjing First Hospital, Nanjing, China
| | - Yuechun Li
- Department of Cardiology, Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yushun Zhang
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hao Hu
- Department of Cardiology, The Second Affiliated Hospital of Lanzhou University, Lanzhou, China
| | - Yawei Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jie Zeng
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Guo
- Department of Cardiology, Senior Department of Cardiology, the Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Xi Su
- Department of Cardiology, Wuhan Asia Heart Hospital, Wuhan, China
| | - Osama Soliman
- Department of Cardiology, National University of Ireland, Galway (NUIG), Galway, Ireland
| | - Patrick W Serruys
- Department of Cardiology, National University of Ireland, Galway (NUIG), Galway, Ireland
| | - Ling Tao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
17
|
Lee SY, Shin MJ, Choi SM, Kim DK, Choi MG, Kim JS, Suh DS, Kim JH, Kim SJ. Role of Peroxisome Proliferator-Activated Receptor α-Dependent Mitochondrial Metabolism in Ovarian Cancer Stem Cells. Int J Mol Sci 2024; 25:11760. [PMID: 39519311 PMCID: PMC11546303 DOI: 10.3390/ijms252111760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/20/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), including PPAR-α, PPAR-β/δ, and PPAR-γ, are involved in various cellular responses, including metabolism and cell proliferation. Increasing evidence suggests that PPARs are closely associated with tumorigenesis and metastasis. However, the exact role of PPARs in energy metabolism and cancer stem cell (CSC) proliferation remains unclear. This study investigated the role of PPARs in energy metabolism and tumorigenesis in ovarian CSCs. The expression of PPARs and fatty acid consumption as an energy source increased in spheroids derived from A2780 ovarian cancer cells (A2780-SP) compared with their parental cells. GW6471, a PPARα inhibitor, induced apoptosis in A2780-SP. PPARα silencing mediated by small hairpin RNA reduced A2780-SP cell proliferation. Treatment with GW6471 significantly inhibited the respiratory oxygen consumption of A2780-SP cells, with reduced dependency on fatty acids, glucose, and glutamine. In a xenograft tumor transplantation mouse model, intraperitoneal injection of GW6471 inhibited in vivo tumor growth of A2780-SP cells. These results suggest that PPARα plays a vital role in regulating the proliferation and energy metabolism of CSCs by altering mitochondrial activity and that it offers a promising therapeutic target to eradicate CSCs.
Collapse
Affiliation(s)
- Seo Yul Lee
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Min Joo Shin
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Seong Min Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Dae Kyoung Kim
- HiCellTech Inc., Yangsan 50612, Gyeongsangnam-do, Republic of Korea;
| | - Mee Gyeon Choi
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Jun Se Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Dong Soo Suh
- Department of Obstetrics and Gynecology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea;
| | - Jae Ho Kim
- Department of Physiology, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea; (S.Y.L.); (M.J.S.); (S.M.C.); (M.G.C.); (J.S.K.)
| | - Seong Jang Kim
- Department of Nuclear Medicine, School of Medicine, Pusan National University, Yangsan 50612, Gyeongsangnam-do, Republic of Korea
| |
Collapse
|
18
|
Iacoboni G, Navarro V, Sesques P, Rejeski K, Bastos-Oreiro M, Serpenti F, Martin Lopez AA, Iraola-Truchuelo J, Delgado J, Perez A, Guerreiro M, Caballero AC, Martinez-Cibrian N, Luzardo Henriquez H, Sanchez Pina JM, Sancho JM, Ghesquieres H, Mussetti A, Lopez Corral L, Hernani R, Reguera JL, Sureda A, Bosch F, Martin Garcia-Sancho A, Kwon M, Subklewe M, Kuhnl A, Bachy E, Barba P, Villacampa G, Abrisqueta P. Development and validation of the post-CAR prognostic index for large B-cell lymphoma patients after CAR-T progression in third or later line treatment. J Hematol Oncol 2024; 17:102. [PMID: 39468591 PMCID: PMC11520873 DOI: 10.1186/s13045-024-01608-8] [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: 07/22/2024] [Accepted: 09/15/2024] [Indexed: 10/30/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy fails to achieve durable responses in over 60% of relapsed/refractory (R/R) large B-cell lymphoma (LBCL) patients in the third or later line setting. After CAR-T failure, survival outcomes are heterogeneous and a prognostic model in this patient population is lacking. A training cohort of 216 patients with progressive disease (PD) after CAR-T from 12 Spanish centers was used to develop the Post-CAR Prognostic Index (PC-PI); primary endpoint was overall survival (OS) from CAR-T progression. Validation was performed in an external cohort from three different European centers (n = 204). The prognostic score incorporated five variables, assessed at time of PD to CAR-T: ECOG (> 0), hemoglobin (< 10 g/dL), LDH (≥ 2xULN), number of extranodal sites (> 1) and time from CAR-T to PD (< 4 months). Patients were classified in four risk groups with distinct OS (p-value < 0.05 in all comparisons). In the validation cohort, median OS in the low (31%), intermediate-low (26%), intermediate-high (17%) and high risk (26%) were 15.7, 7.1, 1.8 and 1.0 months, respectively (p < 0.05 in all comparisons). Results were consistent following adjustment for subsequent treatment. In the external cohort, the PC-PI showed a C-statistic of 0.79 (95%CI 0.76-0.82), outperforming IPI and R-IPI. In conclusion, the PC-PI score is a novel tool for OS prediction and could facilitate risk-adapted management of LBCL patients relapsing after CAR T-cells. Additionally, these results will help stratification and interpretation of trials and real-world data incorporating CART-exposed patients.
Collapse
Affiliation(s)
- Gloria Iacoboni
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain.
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Víctor Navarro
- Statistics Unit, Vall d´Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Kai Rejeski
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Mariana Bastos-Oreiro
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Fabio Serpenti
- Department of Hematology, King's College Hospital, London, UK
| | - Ana Africa Martin Lopez
- Hematology Department, IBSAL, CIBERONC, University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Josu Iraola-Truchuelo
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Javier Delgado
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Seville, Spain
| | - Ariadna Perez
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Manuel Guerreiro
- Hematology Department, Hospital Universitario La Fe, Instituto de Investigación Sanitaria La Fe (IIS-La Fe), Valencia, Spain
| | | | | | - Hugo Luzardo Henriquez
- Department of Hematology, Hospital Universitario de Gran Canaria Doctor Negrín, Islas Canarias, Spain
| | | | - Juan-Manuel Sancho
- Hematology Department, ICO-IJC Hospital Germans Trias i Pujol, Barcelona, Spain
| | | | - Alberto Mussetti
- Clinical Hematology Department, Institut Català d'Oncologia - L'Hospitalet de Llobregat, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Lucia Lopez Corral
- Hematology Department, IBSAL, CIBERONC, University Hospital of Salamanca, University of Salamanca, Salamanca, Spain
| | - Rafael Hernani
- Hematology Department, Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain
| | - Juan Luis Reguera
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), CSIC, Universidad de Sevilla, Seville, Spain
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia - L'Hospitalet de Llobregat, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Francesc Bosch
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marion Subklewe
- Department of Medicine III - Hematology/Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Andrea Kuhnl
- Department of Hematology, King's College Hospital, London, UK
| | | | - Pere Barba
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Pau Abrisqueta
- Department of Hematology, University Hospital Vall d'Hebron, Barcelona, Spain
- Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
19
|
Song YC, Chen SY, Zhao XR, Jing H, Fang H, Tang Y, Hu SY, Song YW, Jin J, Liu YP, Qi SN, Sun GY, Zhong QZ, Du XH, Liu J, Li YX, Wang SL. Prognostic value of lymphocytes in patients with breast cancer receiving radiotherapy after breast-conserving surgery: A post hoc analysis of a phase III randomized trial. Radiother Oncol 2024; 199:110390. [PMID: 38897316 DOI: 10.1016/j.radonc.2024.110390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE To evaluate the prognostic value of peripheral lymphocyte count (PLC) in the breast cancer patients after breast-conserving surgery (BCS) with radiotherapy (RT). METHODS AND MATERIALS This post hoc analysis was performed using data of 628 patients from a phase III, randomized controlled trial comparing hypofractionated RT (HFRT) with conventional fractionated RT (CFRT) after BCS. PLCs were obtained before, during, and after RT until the 1-year follow-up. The optimal cut-off PLCs were determined using the maxstat package in R. Survival rates were estimated using the Kaplan-Meier method and compared with the log-rank test. RESULTS A total of 275 (46.1 %) patients developed lymphopenia during RT, among them, 17 (2.8 %) had grade 3 lymphopenia and no one developed grade 4 lymphopenia. With a median follow-up of 110.8 months, patients with pre-RT PLCs of < 1.77 × 109/L had a significantly lower 10-year breast cancer-specific survival (BCSS) rate (P = 0.013) and overall survival (OS) rate (P = 0.026). Patients with a nadir PLC of < 1.35 × 109/L had a significantly poorer 10-year OS rate (P = 0.048). Multivariate analysis showed that a pre-RT PLC of < 1.77 × 109/L was an independent factor influencing BCSS and OS, while the effect of the nadir PLC did not remain significant. Neither PLC nor lymphopenia recovery at post-RT 1, 3, and 6 months and 1 year was associated with survival. CONCLUSIONS Radiation-induced lymphopenia in patients with breast cancer after BCS tends to be mild. The lower pre-RT PLC predicted poorer survival.
Collapse
Affiliation(s)
- Yu-Chun Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Si-Ye Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Xu-Ran Zhao
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hao Jing
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hui Fang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Tang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shang-Ying Hu
- Department of Cancer Epidemiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Wen Song
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yue-Ping Liu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shu-Nan Qi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guang-Yi Sun
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qiu-Zi Zhong
- Department of Radiation Oncology, Beijing Hospital, Ministry of Health, Beijing, China
| | - Xiang-Hui Du
- Department of Radiation Therapy, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Juan Liu
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Ye-Xiong Li
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Shu-Lian Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
20
|
Dominik N, Balcar L, Semmler G, Simbrunner B, Schwarz M, Hofer BS, Hartl L, Jachs M, Scheiner B, Pinter M, Trauner M, Mandorfer M, Pilger A, Reiberger T. Prevalence and prognostic value of zinc and selenium deficiency in advanced chronic liver disease. Aliment Pharmacol Ther 2024; 60:876-887. [PMID: 39072822 DOI: 10.1111/apt.18179] [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: 02/25/2024] [Revised: 03/24/2024] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND AND AIMS Zinc and selenium are essential trace elements involved in important (patho)physiological processes. The prevalence and prognostic implications of zinc and selenium deficiency in patients with advanced chronic liver disease (ACLD) remain unknown. METHODS We determined serum zinc and selenium concentrations in 309 patients with ACLD undergoing hepatic venous pressure gradient (HVPG) measurement between 2019 and 2022. We evaluated the prevalence of zinc/selenium deficiency and assessed its association with severity of ACLD and liver-related events (LRE, i.e. first/further hepatic decompensation/liver-related death). RESULTS Among 309 ACLD patients (median: age: 57 [IQR: 50-64], MELD: 11 [IQR: 9-16], HVPG: 17 [IQR: 11-20]), 73% (227) and 63% (195) were deficient in zinc and selenium, respectively. Decompensated (dACLD) patients showed significantly lower serum zinc (median: 48 [IQR: 38-59] vs. compensated, cACLD: 65 [IQR: 54-78], p < 0.001) and selenium levels (median: 4.9 [IQR 4.0-6.2] vs. cACLD: 6.1 [IQR 5.1-7.3], p < 0.001). Significant correlations of zinc/selenium levels were found with MELD (zinc: ρ = -0.498, p < 0.001; selenium: ρ = -0.295, p < 0.001), HVPG (zinc: ρ = -0.400, p < 0.001; selenium: ρ = -0.157, p = 0.006) and liver disease-driving mechanisms (IL6, bile-acid homeostasis). On multivariable analysis, low zinc/selenium levels, age and MELD remained independently associated with LRE. CONCLUSION Zinc and selenium deficiencies are common in ACLD patients especially with higher MELD and HVPG. Low zinc and selenium levels independently predicted hepatic decompensation and liver-related death. The effect of zinc/selenium supplementation in ACLD should be investigated in future trials.
Collapse
Affiliation(s)
- Nina Dominik
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lorenz Balcar
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Georg Semmler
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt Simbrunner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Schwarz
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Benedikt S Hofer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mathias Jachs
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bernhard Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Matthias Pinter
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Mattias Mandorfer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Alexander Pilger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
21
|
Zhang N, Yang M, Yang JM, Zhang CY, Guo AY. A Predictive Network-Based Immune Checkpoint Blockade Immunotherapeutic Signature Optimizing Patient Selection and Treatment Strategies. SMALL METHODS 2024; 8:e2301685. [PMID: 38546036 DOI: 10.1002/smtd.202301685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/01/2024] [Indexed: 10/18/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has brought significant advancements to the field of oncology. However, the diverse responses among patients highlight the need for more accurate predictive tools. In this study, insights are drawn from tumor-immunology pathways, and a novel network-based ICB immunotherapeutic signature, termed ICBnetIS, is constructed. The signature is derived from advanced biological network-based computational strategies involving co-expression networks and molecular interactions networks. The efficacy of ICBnetIS is established through its association with enhanced patient survival and a robust immune response characterized by diverse immune cell infiltration and active anti-tumor immune pathways. The validation process positions ICBnetIS as an effective tool in predicting responses to ICB therapy, analyzing ICB data from a broad collection of over 700 samples from multiple cancer types of more than 15 datasets. It achieves an aggregated prediction AUC of 0.784, which outperforms the other nine renowned immunotherapeutic signatures, indicating the superior predictive capability of ICBnetIS. To sum up, the findings suggest ICBnetIS as a potent tool in predicting ICB therapy responses, offering significant implications for patient selection and treatment optimization in oncology. The study highlights the role of ICBnetIS in advancing personalized treatment strategies, potentially transforming the clinical landscape of ICB therapy.
Collapse
Affiliation(s)
- Nan Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Mei Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jing-Min Yang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chu-Yu Zhang
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - An-Yuan Guo
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
22
|
Rodríguez-Arbolí E, Othus M, Freeman SD, Buccisano F, Ngai LL, Thomas I, Palmieri R, Cloos J, Johnson S, Meddi E, Russell NH, Venditti A, Gradowska P, Ossenkoppele GJ, Löwenberg B, Walter RB. Optimal prognostic threshold for measurable residual disease positivity by multiparameter flow cytometry in acute myeloid leukemia (AML). Leukemia 2024; 38:2266-2269. [PMID: 39169114 PMCID: PMC11438566 DOI: 10.1038/s41375-024-02378-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024]
Affiliation(s)
- Eduardo Rodríguez-Arbolí
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Hematology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS/CSIC), University of Seville, Seville, Spain
| | - Megan Othus
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Sylvie D Freeman
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Lok Lam Ngai
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ian Thomas
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Raffaele Palmieri
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Jacqueline Cloos
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sean Johnson
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Elisa Meddi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Adriano Venditti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Gert J Ossenkoppele
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bob Löwenberg
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Roland B Walter
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Wang QY, Zhong WT, Xiao Y, Lin GL, Lu JY, Xu L, Zhang GN, Du JF, Wu B. Pan-immune-inflammation value as a prognostic biomarker for colon cancer and its variation by primary tumor location. World J Gastroenterol 2024; 30:3823-3836. [PMID: 39351432 PMCID: PMC11438628 DOI: 10.3748/wjg.v30.i33.3823] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/02/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
BACKGROUND A growing body of research indicates significant differences between left-sided colon cancers (LCC) and right-sided colon cancers (RCC). Pan-immune-inflammation value (PIV) is a systemic immune response marker that can predict the prognosis of patients with colon cancer. However, the specific distinction between PIV of LCC and RCC remains unclear. AIM To investigate the prognostic and clinical significance of PIV in LCC and RCC patients. METHODS This multicenter retrospective cohort study included 1510 patients with colon cancer, comprising 801 with LCC and 709 with RCC. We used generalized lifting regression analysis to evaluate the relative impact of PIV on disease-free survival (DFS) in these patients. Kaplan-Meier analysis, as well as univariate and multivariate analyses, were used to examine the risk factors for DFS. The correlation between PIV and the clinical characteristics was statistically analyzed in these patients. RESULTS A total of 1510 patients {872 female patients (58%); median age 63 years [interquartile ranges (IQR): 54-71]; patients with LCC 801 (53%); median follow-up 44.17 months (IQR 29.67-62.32)} were identified. PIV was significantly higher in patients with RCC [median (IQR): 214.34 (121.78-386.72) vs 175.87 (111.92-286.84), P < 0.001]. After propensity score matching, no difference in PIV was observed between patients with LCC and RCC [median (IQR): 182.42 (111.88-297.65) vs 189.45 (109.44-316.02); P = 0.987]. PIV thresholds for DFS were 227.84 in LCC and 145.99 in RCC. High PIV (> 227.84) was associated with worse DFS in LCC [PIV-high: Adjusted hazard ratio (aHR) = 2.39; 95% confidence interval: 1.70-3.38; P < 0.001] but not in RCC (PIV-high: aHR = 0.72; 95% confidence interval: 0.48-1.08; P = 0.114). CONCLUSION These findings suggest that PIV may predict recurrence in patients with LCC but not RCC, underscoring the importance of tumor location when using PIV as a colon cancer biomarker.
Collapse
Affiliation(s)
- Qian-Yu Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wen-Tao Zhong
- Medical Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Yi Xiao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Guo-Le Lin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun-Yang Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Lai Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Guan-Nan Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jun-Feng Du
- Medical Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Bin Wu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
24
|
Moradi K, Mohammadi S, Roemer FW, Momtazmanesh S, Hathaway Q, Ibad HA, Hunter DJ, Guermazi A, Demehri S. Progression of Bone Marrow Lesions and the Development of Knee Osteoarthritis: Osteoarthritis Initiative Data. Radiology 2024; 312:e240470. [PMID: 39287521 PMCID: PMC11449232 DOI: 10.1148/radiol.240470] [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: 02/20/2024] [Revised: 06/21/2024] [Accepted: 07/08/2024] [Indexed: 09/19/2024]
Abstract
Background Bone marrow lesions (BMLs) are a known risk factor for incident knee osteoarthritis (OA), and deep learning (DL) methods can assist in automated segmentation and risk prediction. Purpose To develop and validate a DL model for quantifying tibiofemoral BML volume on MRI scans in knees without radiographic OA and to assess the association between longitudinal BML changes and incident knee OA. Materials and Methods This retrospective study included knee MRI scans from the Osteoarthritis Initiative prospective cohort (February 2004-October 2015). The DL model, developed between August and October 2023, segmented the tibiofemoral joint into 10 subregions and measured BML volume in each subregion. Baseline and 4-year follow-up MRI scans were analyzed. Knees without OA at baseline were categorized into three groups based on 4-year BML volume changes: BML-free, BML regression, and BML progression. The risk of developing radiographic and symptomatic OA over 9 years was compared among these groups. Results Included were 3869 non-OA knees in 2430 participants (mean age, 59.5 years ± 9.0 [SD]; female-to-male ratio, 1.3:1). At 4-year follow-up, 2216 knees remained BML-free, 1106 showed an increase in BML volume, and 547 showed a decrease in BML volume. BML progression was associated with a higher risk of developing radiographic knee OA compared with remaining BML-free (hazard ratio [HR] = 3.0; P < .001) or BML regression (HR = 2.0; P < .001). Knees with BML progression also had a higher risk of developing symptomatic OA compared with BML-free knees (HR = 1.3; P < .001). Larger volume changes in BML progression were associated with a higher risk of developing both radiographic OA (HR = 2.0; P < .001) and symptomatic OA (HR = 1.7; P < .001). In almost all subchondral plates, especially the medial femur and tibia, BML progression was associated with a higher risk of developing both radiographic and symptomatic OA compared with remaining BML-free. Conclusion Knees with BML progression, according to subregion and extent of volume changes, were associated with an increased risk of OA compared with BML-free knees and knees with BML regression, highlighting the potential utility of monitoring BML volume changes in evaluating interventions to prevent OA development. ClinicalTrials.gov Identifier: NCT00080171 © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Said and Sakly in this issue.
Collapse
Affiliation(s)
- Kamyar Moradi
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - Soheil Mohammadi
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - Frank W. Roemer
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - Sara Momtazmanesh
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - Quincy Hathaway
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - Hamza Ahmed Ibad
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | - David J. Hunter
- From the Russell H. Morgan Department of Radiology and Radiological
Science, Johns Hopkins University School of Medicine, 601 N Caroline St, JHOC
5165, Baltimore, MD 21287 (K.M., H.A.I., S.D.); Tehran University of Medical
Sciences School of Medicine, Tehran, Iran (S. Mohammadi, S. Momtazmanesh);
Department of Radiology, Boston University Chobanian & Avedisian School
of Medicine, Boston, Mass (F.W.R., A.G.); Department of Radiology,
Universitätsklinikum Erlangen and Friedrich-Alexander-Universität
Erlangen-Nürnberg, Erlangen, Germany (F.W.R.); West Virginia University
School of Medicine, Morgantown, WV (Q.H.); Department of Rheumatology,
University of Sydney, Camperdown, Australia (D.J.H.); and Royal North Shore
Hospital, St. Leonards, Sydney, Australia (D.J.H.)
| | | | | |
Collapse
|
25
|
Tominaga M, Yamazaki M, Umezu H, Sugino H, Fuzawa Y, Yagi T, Ishikawa H. Prognostic Value and Pathological Correlation of Peritumoral Radiomics in Surgically Resected Non-Small Cell Lung Cancer. Acad Radiol 2024; 31:3801-3810. [PMID: 38402002 DOI: 10.1016/j.acra.2024.01.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/24/2024] [Accepted: 01/24/2024] [Indexed: 02/26/2024]
Abstract
RATIONALE AND OBJECTIVES To determine the additional value of peritumoral radiomics in predicting overall survival (OS) in surgically resected non-small cell lung cancer (NSCLC) and its correlation with pathological findings. METHODS A total of 526 patients with surgically resected NSCLC were included (191 training, 160 internal validation, and 175 external validation cohorts). CT images were used to segment the gross tumor volume (GTV) and peritumoral volume (PTV) within distances of 3, 6, 9 mm from the tumor boundary (PTV3, PTV6, and PTV9), and radiomic features were extracted. Four prognostic models for OS (GTV, GTV + PTV3, GTV + PTV6, and GTV + PTV9) were constructed using the training cohort. The prognostic ability and feature importance were evaluated using the validation cohorts. Pathological findings were compared between the two patient groups (n = 30 for each) having the top 30 and bottom 30 values of the most important peritumoral feature. RESULTS The GTV+ PTV3 models exhibited the highest predictive ability, which was higher than that of the GTV model in the internal validation cohort (C-index: 0.666 vs. 0.616, P = 0.027) and external validation cohort (C-index: 0.705 vs. 0.656, P = 0.048). The most important feature was GLDM_Dependence_Entropy, extracted from PTV3. High peritumoral GLDM_Dependence_Entropy was associated with a high proportion of invasive histological types, tumor spread through air spaces, and tumor-infiltrating lymphocytes (all P < 0.05). CONCLUSION The GTV and PTV3 combination demonstrated a higher prognostic ability, compared to GTV alone. Peritumoral radiomic features may be associated with various pathological prognostic factors.
Collapse
Affiliation(s)
- Masaki Tominaga
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Motohiko Yamazaki
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Hajime Umezu
- Division of Pathology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Hideaki Sugino
- Division of Pathology, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Yuma Fuzawa
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takuya Yagi
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Ishikawa
- Department of Radiology and Radiation Oncology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
26
|
Würstle S, Schneider T, Karapetyan S, Hapfelmeier A, Isaakidou A, Studen F, Schmid RM, von Delius S, Rothe K, Burgkart R, Obermeier A, Triebelhorn J, Erber J, Voit F, Geisler F, Spinner CD, Schneider J, Wagner L. LINAS-Score: prognostic model for mortality assessment in patients with cirrhotic liver and infected ascites. J Gastroenterol Hepatol 2024; 39:1876-1884. [PMID: 38837839 DOI: 10.1111/jgh.16637] [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: 11/25/2023] [Revised: 04/21/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND AND AIM Patients with liver cirrhosis often face a grave threat from infected ascites (IA). However, a well-established prognostic model for this complication has not been established in routine clinical practice. Therefore, we aimed to assess mortality risk in patients with liver cirrhosis and IA. METHODS We conducted a retrospective study across three tertiary hospitals, enrolling 534 adult patients with cirrhotic liver and IA, comprising 465 with spontaneous bacterial peritonitis (SBP), 34 with bacterascites (BA), and 35 with secondary peritonitis (SP). To determine the attributable mortality risk linked to IA, these patients were matched with 122 patients with hydropic decompensated liver cirrhosis but without IA. Clinical, laboratory, and microbiological parameters were assessed for their relation to mortality using univariable analyses and a multivariable random forest model (RFM). Least absolute shrinkage and selection operator (Lasso) regression model was used to establish an easy-to-use mortality prediction score. RESULTS The in-hospital mortality risk was highest for SP (39.0%), followed by SBP (26.0%) and BA (25.0%). Besides illness severity markers, microbiological parameters, such as Candida spp., were identified as the most significant indicators for mortality. The Lasso model determined 15 parameters with corresponding scores, yielding good discriminatory power (area under the receiver operating characteristics curve = 0.89). Counting from 0 to 83, scores of 20, 40, 60, and 80 corresponded to in-hospital mortalities of 3.3%, 30.8%, 85.2%, and 98.7%, respectively. CONCLUSION We developed a promising mortality prediction score for IA, highlighting the importance of microbiological parameters in conjunction with illness severity for assessing patient outcomes.
Collapse
Affiliation(s)
- Silvia Würstle
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
- Department of Internal Medicine, Infectious Diseases, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tillman Schneider
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Siranush Karapetyan
- TUM School of Medicine and Health, Department of General Practice and Health Services Research, University Medical Center, Technical University of Munich, Munich, Germany
- TUM School of Medicine and Health, Department of AI and Informatics in Medicine, University Medical Center, Technical University of Munich, Munich, Germany
| | - Alexander Hapfelmeier
- TUM School of Medicine and Health, Department of General Practice and Health Services Research, University Medical Center, Technical University of Munich, Munich, Germany
- TUM School of Medicine and Health, Department of AI and Informatics in Medicine, University Medical Center, Technical University of Munich, Munich, Germany
| | - Andriana Isaakidou
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Fabian Studen
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Roland M Schmid
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Stephan von Delius
- Department of Internal Medicine II, RoMed Hospital Rosenheim, Rosenheim, Germany
| | - Kathrin Rothe
- TUM School of Medicine and Health, Department of Medical Microbiology, Immunology and Hygiene, University Medical Center, Technical University of Munich, Munich, Germany
| | - Rainer Burgkart
- TUM School of Medicine and Health, Department of Orthopaedics and Sports Orthopaedics, University Medical Center, Technical University of Munich, Munich, Germany
| | - Andreas Obermeier
- TUM School of Medicine and Health, Department of Orthopaedics and Sports Orthopaedics, University Medical Center, Technical University of Munich, Munich, Germany
| | - Julian Triebelhorn
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Johanna Erber
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Florian Voit
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Fabian Geisler
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| | - Christoph D Spinner
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Jochen Schneider
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
- German Centre for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Laura Wagner
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, Munich, Germany
| |
Collapse
|
27
|
Lv J, Xu LX, Li ZX, Lin L, Wu CF, Quan TQ, Zhen ZC, Li WF, Tang LL, Mao YP, Chen L, Guo R, Zhang LL, Ai XL, Wu SY, Hao MY, Wei D, Li JB, Ma J, Chen YP, Zhou GQ, Sun Y. Longitudinal on-treatment circulating tumor DNA as a biomarker for real-time dynamic risk monitoring in cancer patients: The EP-SEASON study. Cancer Cell 2024; 42:1401-1414.e4. [PMID: 39059389 DOI: 10.1016/j.ccell.2024.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Recurrence risks of cancer patient can change during treatment as a result of treatment-related tumor evolution. However, biomarkers that can monitor these changes are lacking. Here, we investigated whether tracking circulating tumor DNA (ctDNA) dynamics through liquid biopsy can inform real-time recurrence risk. Nasopharyngeal carcinoma (NPC) provides an ideal model where cell-free Epstein-Barr virus (EBV) DNA (cfEBV DNA), a ctDNA, can be sensitively detected. We conducted the EP-SEASON study (NCT03855020) and prospectively recruited 1,000 NPC patients undergoing per-protocol cfEBV DNA assessments at 11 time points and receiving sequential chemo-radiotherapy. Longitudinal cfEBV DNA displayed distinct patterns during neoadjuvant chemotherapy and radiotherapy. Despite the prognostic significance of cfEBV DNA at each time point, real-time recurrence risks changed in sync with cfEBV DNA dynamics. Furthermore, we identified phenotypes of whole-course ctDNA changing dynamics associated with different survival outcomes. In conclusion, tracking longitudinal on-treatment ctDNA can forecast real-time recurrence risk, facilitating risk-adapted, individualized patient management.
Collapse
Affiliation(s)
- Jiawei Lv
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ling-Xin Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Zhi-Xuan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Li Lin
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Chen-Fei Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ting-Qiu Quan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Zi-Cheng Zhen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Wen-Fei Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ling-Long Tang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Yan-Ping Mao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Lei Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Rui Guo
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Lu-Lu Zhang
- Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin-Lei Ai
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Shi-Yue Wu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Meng-Yu Hao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Denghui Wei
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China
| | - Ji-Bin Li
- Clinical Trial Centre, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun Ma
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Yu-Pei Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Guan-Qun Zhou
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| | - Ying Sun
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Department of Radiation Oncology, Guangzhou 510060, China.
| |
Collapse
|
28
|
Talarico MCR, Derchain S, da Silva LF, Sforça ML, Rocco SA, Cardoso MR, Sarian LO. Metabolomic Profiling of Breast Cancer Patients Undergoing Neoadjuvant Chemotherapy for Predicting Disease-Free and Overall Survival. Int J Mol Sci 2024; 25:8639. [PMID: 39201325 PMCID: PMC11354796 DOI: 10.3390/ijms25168639] [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: 06/27/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Breast cancer (BC) remains a significant global health concern, with neoadjuvant chemotherapy (NACT) offering preoperative benefits like tumor downstaging and treatment response assessment. However, identifying factors influencing post-NACT treatment response and survival outcomes is challenging. Metabolomic approaches offer promising insights into understanding these outcomes. This study analyzed the serum of 80 BC patients before and after NACT, followed for up to five years, correlating with disease-free survival (DFS) and overall survival (OS). Using untargeted nuclear magnetic resonance (NMR) spectroscopy and a novel statistical model that avoids collinearity issues, we identified metabolic changes associated with survival outcomes. Four metabolites (histidine, lactate, serine, and taurine) were significantly associated with DFS. We developed a metabolite-related survival score (MRSS) from these metabolites, stratifying patients into low- and high-risk relapse groups, independent of classical prognostic factors. High-risk patients had a hazard ratio (HR) for DFS of 3.42 (95% CI 1.51-7.74; p = 0.003) after adjustment for disease stage and age. A similar trend was observed for OS (HR of 3.34, 95% CI 1.64-6.80; p < 0.001). Multivariate Cox proportional hazards analysis confirmed the independent prognostic value of the MRSS. Our findings suggest the potential of metabolomic data, alongside traditional markers, in guiding personalized treatment decisions and risk stratification in BC patients undergoing NACT. This study provides a methodological framework for leveraging metabolomics in survival analyses.
Collapse
Affiliation(s)
- Maria Cecília Ramiro Talarico
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| | - Sophie Derchain
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| | | | - Maurício L. Sforça
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Silvana A. Rocco
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Marcella R. Cardoso
- Division of Gynecologic Oncology-MGH Global Disaster Response, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Center for Global Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luís Otávio Sarian
- Department of Obstetrics and Gynecology, Division of Gynecologic and Breast Oncology, School of Medical Sciences, University of Campinas (UNICAMP-Universidade Estadual de Campinas), Campinas 13083-881, SP, Brazil
| |
Collapse
|
29
|
Khalil C, Khoury M, Higgins K, Enepekides D, Karam I, Husain ZA, Bayley A, Poon I, Truong T, Chan KKW, Smoragiewicz M, Fu R, Eskander A. Lymph node yield: Impact on oncologic outcomes in oral cavity cancer. Head Neck 2024; 46:1965-1974. [PMID: 38344842 DOI: 10.1002/hed.27656] [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: 07/19/2023] [Revised: 01/04/2024] [Accepted: 01/14/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Lymph node metastases are associated with poor prognosis in oral cavity squamous cell carcinoma (OCSCC). In other cancers, clinical guidelines on the number of lymph nodes removed during primary surgery, lymph node yield (LNY), exist. Here, we evaluated the prognostic capacity of LNY on regional failure, locoregional recurrence, and disease-free survival (DFS) in patients with OCSCC treated by primary neck surgery. METHODS This retrospective cohort study took place at Sunnybrook Health Sciences Centre in Toronto, Canada and involved a chart review of all adult patients with treatment-naive OCSCC undergoing primary neck dissection. For each outcome, we first used the maximally selected rank statistics and an optimism-corrected concordance to identify an optimal threshold of LNY. We then used a multivariable Cox proportional hazards model to assess the association between high LNY (>threshold) and each outcome. RESULTS Among the 579 patients with OCSCC receiving primary neck dissection, 61.7% (n = 357) were male with a mean age of 62.9 years (standard deviation: 13.1) at cancer diagnosis. When adjusting for sociodemographic and clinical factors, LNY >15 was significantly associated with improved DFS (adjusted HR [aHR]: 0.73, 95% CI: 0.54-0.98), locoregional recurrence (aHR: 0.68, 95% CI: 0.49-0.95), and regional failure (aHR: 0.61, 95% CI: 0.39-0.93). CONCLUSIONS Our study findings suggested high LNY to be a strong independent predictor of various patient-level quality of surgical care metrics. The optimal LNY we found (15) was lower than the conventionally recommended (18), which calls for further research to establish validity in practice.
Collapse
Affiliation(s)
- Carlos Khalil
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mark Khoury
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kevin Higgins
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Danny Enepekides
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Irene Karam
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Zain Ali Husain
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
- Department of Radiation Oncology, Stanford University, Stanford, California, USA
| | - Andrew Bayley
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Ian Poon
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Tra Truong
- Department of Anatomic Pathology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Kelvin K W Chan
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Martin Smoragiewicz
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Rui Fu
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Antoine Eskander
- Department of Otolaryngology - Head and Neck Surgery, University of Toronto, Toronto, Ontario, Canada
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
- Institute of Health Policy, Management, and Evaluation, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Clerico M, Ferrero S, Alessandria B, Zaccaria GM, Genuardi E, Ragaini S, Tavarozzi R, Cavallo F, Hohaus S, Musuraca G, Carella AM, Stelitano C, Tani M, Gaidano G, Olivieri J, Usai SV, Galimberti S, Re F, Mian M, Castellino C, Pavone V, Evangelista A, Bruno B, Cortelazzo S, Passera R, Ladetto M. Stem cell collection and hematological recovery in the Fondazione Italiana Linfomi (FIL) MCL0208 clinical trial. Sci Rep 2024; 14:16946. [PMID: 39043871 PMCID: PMC11266400 DOI: 10.1038/s41598-024-67906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
In the frontline high-dose phase 3 FIL-MCL0208 trial (NCT02354313), 8% of enrolled mantle cell lymphoma (MCL) patients could not be randomised to receive lenalidomide (LEN) maintenance vs observation after autologous stem cell transplantation (ASCT) due to inadequate hematological recovery and 52% of those who started LEN, needed a dose reduction due to toxicity. We therefore focused on the role played by CD34 + hematopoietic stem cells (PBSC) harvesting and reinfusion on toxicity and outcome. Overall, 90% (n = 245) of enrolled patients who underwent the first leukapheresis collected ≥ 4 × 106 PBSC/kg, 2.6% (n = 7) mobilized < 4 × 106 PBSC/kg and 7.7% (n = 21) failed the collection. Similar results were obtained for the planned second leukapheresis, with only one patient failing both attempts. Median count of reinfused PBSC was 5 × 106/kg and median time to recovery from neutropenia G4 was 10 days from ASCT. No impact of mobilizing subtype or number of reinfused PBSC on hematological recovery and LEN dose reduction was noted. At a median follow-up of 75 months from ASCT, PFS and OS of transplanted patients were 50% and 73%, respectively. A long lasting G4 neutropenia after ASCT (> 10 days) was associated with a worse outcome, both in terms of PFS and OS. In conclusion, although the harvesting procedures proved feasible for younger MCL patients, long-lasting cytopenia following ASCT remains a significant issue: this can hinder the administration of effective maintenance therapies, potentially increasing the relapse rate and negatively affecting survival outcomes.
Collapse
Affiliation(s)
- Michele Clerico
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
| | - Simone Ferrero
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy.
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy.
| | - Beatrice Alessandria
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Gian Maria Zaccaria
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Bari, Italy
| | - Elisa Genuardi
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Simone Ragaini
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Rita Tavarozzi
- SC Ematologia Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Federica Cavallo
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | - Stefan Hohaus
- Institute of Hematology, Università Cattolica del Sacro Cuore and Fondazione Policlinico Univeristario A. Gemelli, Roma, Italy
| | - Gerardo Musuraca
- IRCCS, IRST Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori "Dino Amadori", Meldola, Italy
| | - Angelo Michele Carella
- Department of Hematology and Bone Marrow Transplant, IRCSS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Caterina Stelitano
- Department of Hematology, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Monica Tani
- UOC di Ematologia, Ospedale S. Maria delle Croci, Ravenna, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Jacopo Olivieri
- Clinica Ematologica, Azienda Sanitaria Universitaria Integrata di Udine, Udine, Italy
| | | | - Sara Galimberti
- Division of Hematology, University Hospital of Pisa, Pisa, Italy
| | - Francesca Re
- Division of Immuno-Haematology, AOU Parma, Parma, Italy
| | - Michael Mian
- Innovation, Research and Teaching Service (SABES-ASDAA), Teaching Hospital of the Paracelsus Medical Private University (PMU), Bolzano, Italy
- College of Health Care-Professions Claudiana, Bozen, Italy
| | - Claudia Castellino
- Division of Hematology, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
| | - Vincenzo Pavone
- Ospedale Pia Fondazione Cardinale Panico, Tricase, Lecce, Italy
| | - Andrea Evangelista
- Unit of Clinical Epidemiology, AOU "Città della Salute e della Scienza di Torino" and CPO Piemonte, Torino, Italy
| | - Benedetto Bruno
- Division of Hematology, AOU "Città della Salute e della Scienza di Torino", Torino, Italy
- Department of Molecular Biotechnologies and Health Sciences, University of Torino, Torino, Italy
| | | | - Roberto Passera
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Marco Ladetto
- SC Ematologia Azienda Ospedaliera Santi Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| |
Collapse
|
31
|
Li W, Pan L, Hong W, Ginhoux F, Zhang X, Xiao C, Li X. A single-cell pan-cancer analysis to show the variability of tumor-infiltrating myeloid cells in immune checkpoint blockade. Nat Commun 2024; 15:6142. [PMID: 39034339 PMCID: PMC11271490 DOI: 10.1038/s41467-024-50478-8] [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: 12/03/2023] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
Myeloid cells are vital components of the immune system and have pivotal functions in orchestrating immune responses. Understanding their functions within the tumor microenvironment and their interactions with tumor-infiltrating lymphocytes presents formidable challenges across diverse cancer types, particularly with regards to cancer immunotherapies. Here, we explore tumor-infiltrating myeloid cells (TIMs) by conducting a pan-cancer analysis using single-cell transcriptomics across eight distinct cancer types, encompassing a total of 192 tumor samples from 129 patients. By examining gene expression patterns and transcriptional activities of TIMs in different cancer types, we discern notable alterations in abundance of TIMs and kinetic behaviors prior to and following immunotherapy. We also identify specific cell-cell interaction targets in immunotherapy; unique and shared regulatory profiles critical for treatment response; and TIMs associated with survival outcomes. Overall, our study illuminates the heterogeneity of TIMs and improves our understanding of tissue-specific and cancer-specific myeloid subsets within the context of tumor immunotherapies.
Collapse
Affiliation(s)
- Weiyuan Li
- School of Medicine, Yunnan University, Kunming, Yunnan, 650091, China
- Department of Reproductive Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan, 650032, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650031, China
| | - Lu Pan
- Institute of Environmental Medicine, Karolinska Institutet, Solna, 171 65, Sweden
| | - Weifeng Hong
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, 310005, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310005, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, 310005, China
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, 138648, Singapore
- Institut Gustave Roussy, INSERM U1015, Bâtiment de Médecine Moléculaire 114 rue Edouard Vaillant, 94800, Villejuif, France
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuan Zhang
- Department of Colorectal Surgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Chunjie Xiao
- School of Medicine, Yunnan University, Kunming, Yunnan, 650091, China.
| | - Xuexin Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, 110122, Liaoning, China.
- Institute of Health Sciences, China Medical University, Shenyang, 110122, Liaoning, China.
- Department of Physiology and Pharmacology, Karolinska Institute, Solna, 171 65, Sweden.
| |
Collapse
|
32
|
Cullaro G, Chiou SH, Fenton C, Ge J, McCulloch CE, Rubin J, Shui AM, Yao F, Lai JC. Outpatient mean arterial pressure: A potentially modifiable risk for acute kidney injury and death among patients with cirrhosis. Liver Transpl 2024; 30:679-688. [PMID: 38535488 PMCID: PMC11792088 DOI: 10.1097/lvt.0000000000000359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
Mean arterial blood pressure (MAP), which decreases as portal hypertension progresses, may be a modifiable risk factor among patients with cirrhosis. We included adults enrolled in the Functional Assessment in Liver Transplantation study. We completed latent class trajectory analyses to define MAP trajectories. We completed time-dependent Cox-regression analyses to test the association between outpatient MAP and 3 cirrhosis-related outcomes: (1) stage 2 acute kidney injury (AKI), defined as a ≥200% increase in serum creatinine from baseline; (2) a 5-point increase in the MELD-Na score, defined as the incidence of increase from initial MELD-Na; (3) waitlist mortality, defined as death on the waitlist. For each outcome, we defined MAP cut points by determining the maximally selected Log-rank statistic after univariable Cox-regression analyses. Among the 1786 patients included in this analysis, our latent class trajectory analyses identified 3 specific outpatient MAP trajectories: "stable-low," "stable-high," and "increasing-to-decreasing." However, >80% of patients were in a "stable-low" trajectory. We found in adjusted analyses that outpatient MAP was associated with each of our outcomes: Stage 2 AKI (adjusted hazard ratio 0.88 per 10 mm Hg increase in MAP [95% CI: 0.79-0.99]); 5-point increase in MELD-Na (adjusted hazard ratio: 0.91 [95% CI: 0.86-0.96]; waitlist mortality (adjusted hazard ratio: 0.89 [95% CI: 0.81-0.96]). For each outcome, we found that an outpatient MAP of 82 mm Hg was most associated with outcomes ( p <0.05 for all). Our study informs the association between outpatient MAP and cirrhosis-related outcomes. These findings, coupled with the identification of specific thresholds, lay the foundation for the trial of targeted outpatient MAP modulation in patients with cirrhosis.
Collapse
Affiliation(s)
- Giuseppe Cullaro
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Sy Han Chiou
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Cynthia Fenton
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Jin Ge
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Charles E. McCulloch
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Jessica Rubin
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Amy M. Shui
- Department of Epidemiology and Biostatistics, University of California, San Francisco, California, USA
| | - Frederick Yao
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Jennifer C. Lai
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
33
|
Hua X, Xu F, Shi W, Long ZQ, Huang X, Duan FF, Wang SF, Zhang C, Wang MD, Ni WQ, Xia W, Chen JY, Gao YS. Prognostic significance of platelet‑to‑albumin ratio in patients with nasopharyngeal carcinoma receiving concurrent chemoradiotherapy: a retrospective study of 858 cases. BMC Cancer 2024; 24:762. [PMID: 38918690 PMCID: PMC11197365 DOI: 10.1186/s12885-024-12499-w] [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: 03/27/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Despite evidence supporting the high correlation of the novel platelet-to-albumin ratio (PAR) with survival in diverse malignancies, its prognostic relevance in nasopharyngeal carcinoma (NPC) remains underexplored. This study aimed to examine the link between PAR and overall survival (OS) in NPC and to establish a predictive model based on this biomarker. METHODS We retrospectively assembled a cohort consisting of 858 NPC patients who underwent concurrent chemoradiotherapy (CCRT). Utilizing the maximally selected log-rank method, we ascertained the optimal cut-off point for the PAR. Subsequently, univariate and multivariate Cox proportional hazards models were employed to discern factors significantly associated with OS and to construct a predictive nomogram. Further, we subjected the nomogram's predictive accuracy to rigorous independent validation. RESULTS The discriminative optimal PAR threshold was determined to be 4.47, effectively stratifying NPC patients into two prognostically distinct subgroups (hazard ratio [HR] = 0.53; 95% confidence interval [CI]: 0.28-0.98, P = 0.042). A predictive nomogram was formulated using the results from multivariate analysis, which revealed age greater than 45 years, T stage, N stage, and PAR score as independent predictors of OS. The nomogram demonstrated a commendable predictive capability for OS, with a C-index of 0.69 (95% CI: 0.64-0.75), surpassing the performance of the conventional staging system, which had a C-index of 0.56 (95% CI: 0.65-0.74). CONCLUSIONS In the context of NPC patients undergoing CCRT, the novel nutritional-inflammatory biomarker PAR emerges as a promising, cost-efficient, easily accessible, non-invasive, and potentially valuable predictor of prognosis. The predictive efficacy of the nomogram incorporating the PAR score exceeded that of the conventional staging approach, thereby indicating its potential as an enhanced prognostic tool in this clinical setting.
Collapse
Affiliation(s)
- Xin Hua
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China
| | - Fei Xu
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China
| | - Wei Shi
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Qing Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Xin Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Fang-Fang Duan
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Si-Fen Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Chao Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Meng-Di Wang
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China
| | - Wei-Qiong Ni
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China
| | - Wen Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, SunYat-sen University Cancer Center, Guangzhou, China
| | - Jia-Yi Chen
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China
| | - Yun-Sheng Gao
- Department of Radiation Oncology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, Shanghai, China.
| |
Collapse
|
34
|
Castaneda CA, Castillo M, Bernabe LA, Sanchez J, Fassan M, Tello K, Wistuba II, Chavez Passiuri I, Ruiz E, Sanchez J, Barreda F, Valdivia D, Bazan Y, Abad-Licham M, Mengoa C, Fuentes H, Montenegro P, Poquioma E, Alatrista R, Flores CJ, Taxa L. Association between Helicobacter pylori infection, mismatch repair, HER2 and tumor-infiltrating lymphocytes in gastric cancer. World J Gastrointest Oncol 2024; 16:2487-2503. [PMID: 38994161 PMCID: PMC11236231 DOI: 10.4251/wjgo.v16.i6.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The influence of Helicobacter-pylori (H. pylori) infection and the characteristics of gastric cancer (GC) on tumor-infiltrating lymphocyte (TIL) levels has not been extensively studied. Analysis of infiltrating-immune-cell subtypes as well as survival is necessary to obtain comprehensive information. AIM To determine the rates of deficient mismatch-repair (dMMR), HER2-status and H. pylori infection and their association with TIL levels in GC. METHODS Samples from 503 resected GC tumors were included and TIL levels were evaluated following the international-TILs-working-group recommendations with assessment of the intratumoral (IT), stromal (ST) and invasive-border (IB) compartments. The density of CD3, CD8 and CD163 immune cells, and dMMR and HER2-status were determined by immunohistochemistry (IHC). H. pylori infection was evaluated by routine histology and quantitative PCR (qPCR) in a subset of samples. RESULTS dMMR was found in 34.4%, HER2+ in 5% and H. pylori-positive in 55.7% of samples. High IT-TIL was associated with grade-3 (P = 0.038), while ST-TIL with grade-1 (P < 0.001), intestinal-histology (P < 0.001) and no-recurrence (P = 0.003). dMMR was associated with high TIL levels in the ST (P = 0.019) and IB (P = 0.01) compartments, and ST-CD3 (P = 0.049) and ST-CD8 (P = 0.05) densities. HER2- was associated with high IT-CD8 (P = 0.009). H. pylori-negative was associated with high IT-TIL levels (P = 0.009) when assessed by routine-histology, and with high TIL levels in the 3 compartments (P = 0.002-0.047) and CD8 density in the IT and ST compartments (P = 0.001) when assessed by qPCR. A longer overall survival was associated with low IT-CD163 (P = 0.003) and CD8/CD3 (P = 0.001 in IT and P = 0.002 in ST) and high IT-CD3 (P = 0.021), ST-CD3 (P = 0.003) and CD3/CD163 (P = 0.002). CONCLUSION TIL levels were related to dMMR and H. pylori-negativity. Low CD8/CD3 and high CD163/CD3 were associated with lower recurrence and longer survival.
Collapse
Affiliation(s)
- Carlos A Castaneda
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima 15038, Peru
- GECO PERU, Grupo de Estudios Clinicos Oncologicos del Peru, Lima 15038, Peru
| | - Miluska Castillo
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Luis A Bernabe
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Joselyn Sanchez
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- Faculty of Human Medicine, Universidad Ricardo Palma, Lima 15039, Peru
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua 35121, Italy
| | - Katherine Tello
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Ignacio Ivan Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Ivan Chavez Passiuri
- Department of Abdominal Surgery, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Eloy Ruiz
- Department of Abdominal Surgery, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Juvenal Sanchez
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Fernando Barreda
- Department of Medical Specialties, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Daniel Valdivia
- Department of Medical Specialties, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Yaqueline Bazan
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Milagros Abad-Licham
- Department of Pathology, Instituto Regional de Enfermedades Neoplasicas del Norte, Trujillo 13001, Peru
- Faculty of Human Medicine, Universidad Privada Antenor Orrego, Trujillo 13008, Peru
| | - Claudio Mengoa
- Department of Surgery, Instituto Regional de Enfermedades Neoplasicas del Sur, Arequipa 04002, Peru
| | - Hugo Fuentes
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Paola Montenegro
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Ebert Poquioma
- Department of Epidemiology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Raul Alatrista
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Claudio J Flores
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15038, Peru
| | - Luis Taxa
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- Faculty of Medicine, Universidad San Martin de Porres, Lima 15008, Peru
| |
Collapse
|
35
|
Castaneda CA, Castillo M, Bernabe LA, Sanchez J, Fassan M, Tello K, Wistuba II, Chavez Passiuri I, Ruiz E, Sanchez J, Barreda F, Valdivia D, Bazan Y, Abad-Licham M, Mengoa C, Fuentes H, Montenegro P, Poquioma E, Alatrista R, Flores CJ, Taxa L. Association between Helicobacter pylori infection, mismatch repair, HER2 and tumor-infiltrating lymphocytes in gastric cancer. World J Gastrointest Oncol 2024; 16:2475-2491. [DOI: 10.4251/wjgo.v16.i6.2475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND The influence of Helicobacter-pylori (H. pylori) infection and the characteristics of gastric cancer (GC) on tumor-infiltrating lymphocyte (TIL) levels has not been extensively studied. Analysis of infiltrating-immune-cell subtypes as well as survival is necessary to obtain comprehensive information.
AIM To determine the rates of deficient mismatch-repair (dMMR), HER2-status and H. pylori infection and their association with TIL levels in GC.
METHODS Samples from 503 resected GC tumors were included and TIL levels were evaluated following the international-TILs-working-group recommendations with assessment of the intratumoral (IT), stromal (ST) and invasive-border (IB) compartments. The density of CD3, CD8 and CD163 immune cells, and dMMR and HER2-status were determined by immunohistochemistry (IHC). H. pylori infection was evaluated by routine histology and quantitative PCR (qPCR) in a subset of samples.
RESULTS dMMR was found in 34.4%, HER2+ in 5% and H. pylori-positive in 55.7% of samples. High IT-TIL was associated with grade-3 (P = 0.038), while ST-TIL with grade-1 (P < 0.001), intestinal-histology (P < 0.001) and no-recurrence (P = 0.003). dMMR was associated with high TIL levels in the ST (P = 0.019) and IB (P = 0.01) compartments, and ST-CD3 (P = 0.049) and ST-CD8 (P = 0.05) densities. HER2- was associated with high IT-CD8 (P = 0.009). H. pylori-negative was associated with high IT-TIL levels (P = 0.009) when assessed by routine-histology, and with high TIL levels in the 3 compartments (P = 0.002-0.047) and CD8 density in the IT and ST compartments (P = 0.001) when assessed by qPCR. A longer overall survival was associated with low IT-CD163 (P = 0.003) and CD8/CD3 (P = 0.001 in IT and P = 0.002 in ST) and high IT-CD3 (P = 0.021), ST-CD3 (P = 0.003) and CD3/CD163 (P = 0.002).
CONCLUSION TIL levels were related to dMMR and H. pylori-negativity. Low CD8/CD3 and high CD163/CD3 were associated with lower recurrence and longer survival.
Collapse
Affiliation(s)
- Carlos A Castaneda
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima 15038, Peru
- GECO PERU, Grupo de Estudios Clinicos Oncologicos del Peru, Lima 15038, Peru
| | - Miluska Castillo
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Luis A Bernabe
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Joselyn Sanchez
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- Faculty of Human Medicine, Universidad Ricardo Palma, Lima 15039, Peru
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua 35121, Italy
| | - Katherine Tello
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Ignacio Ivan Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Ivan Chavez Passiuri
- Department of Abdominal Surgery, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Eloy Ruiz
- Department of Abdominal Surgery, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Juvenal Sanchez
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Fernando Barreda
- Department of Medical Specialties, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Daniel Valdivia
- Department of Medical Specialties, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Yaqueline Bazan
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Milagros Abad-Licham
- Department of Pathology, Instituto Regional de Enfermedades Neoplasicas del Norte, Trujillo 13001, Peru
- Faculty of Human Medicine, Universidad Privada Antenor Orrego, Trujillo 13008, Peru
| | - Claudio Mengoa
- Department of Surgery, Instituto Regional de Enfermedades Neoplasicas del Sur, Arequipa 04002, Peru
| | - Hugo Fuentes
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Paola Montenegro
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Ebert Poquioma
- Department of Epidemiology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Raul Alatrista
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
| | - Claudio J Flores
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima 15038, Peru
| | - Luis Taxa
- Department of Pathology, Instituto Nacional de Enfermedades Neoplasicas, Lima 15038, Peru
- Faculty of Medicine, Universidad San Martin de Porres, Lima 15008, Peru
| |
Collapse
|
36
|
Zhou E, Wu J, Zhou X, Yin Y. Systemic inflammatory biomarkers are novel predictors of all-cause and cardiovascular mortality in individuals with osteoarthritis: a prospective cohort study using data from the NHANES. BMC Public Health 2024; 24:1586. [PMID: 38872115 PMCID: PMC11170786 DOI: 10.1186/s12889-024-19105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 06/11/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Chronic inflammation may contribute to increased mortality risk in individuals with osteoarthritis (OA), but research on the prognostic value of inflammatory biomarkers is limited. We aimed to evaluate the associations of the systemic immune-inflammation index (SII) and systemic inflammation response index (SIRI) with all-cause and cardiovascular mortality among US adults with OA. METHODS This cohort study included 3545 adults with OA aged ≥ 20 years from the National Health and Nutrition Examination Survey 1999-2020. The SII and SIRI were calculated using complete blood cell count data. Participants were categorized as having a higher or lower SII and SIRI using cutoff points derived by the maximally selected rank statistics method. Cox proportional hazards models, Fine-Gray competing risk regression models and time-dependent receiver operating characteristic (ROC) analysis were used to evaluate the associations between the SII/SIRI and mortality in OA patients. RESULTS Over a median follow-up of 5.08 (3.42-9.92) years, 636 (17.94%) deaths occurred, including 149 (4.20%) cardiovascular deaths. According to multivariable-adjusted models involving demographic, socioeconomic, and health factors, OA patients with a higher SII had a twofold greater risk of all-cause mortality than patients with a lower SII (HR 2.01; 95% CI: 1.50-2.68). Similarly, a higher SIRI was associated with an 86% increased risk of all-cause mortality relative to a lower SIRI (HR 1.86; 95% CI: 1.46-2.38). Similar to the trend found with all-cause mortality, patients with an elevated SII and SIRI had a 88% and 67% increased risk of cardiovascular mortality, respectively, compared to patients with a lower SII (HR 1.88; 95% CI: 1.16-3.03) and SIRI (HR 1.67; 95% CI: 1.14-2.44). Time-dependent ROC curves showed that both the SII and SIRI have moderate and valid performance in predicting short- and long-term mortality in patients with OA. CONCLUSIONS Higher SII and SIRI values were associated with greater all-cause and cardiovascular mortality among US adults with OA.
Collapse
Affiliation(s)
- Erye Zhou
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou , Jiangsu, 215006, China
| | - Jian Wu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou , Jiangsu, 215006, China
| | - Xin Zhou
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou , Jiangsu, 215006, China
| | - Yufeng Yin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Soochow University, No.188 Shizi St, Suzhou , Jiangsu, 215006, China.
| |
Collapse
|
37
|
Pilcher WC, Yao L, Gonzalez-Kozlova E, Pita-Juarez Y, Karagkouni D, Acharya CR, Michaud ME, Hamilton M, Nanda S, Song Y, Sato K, Wang JT, Satpathy S, Ma Y, Schulman J, D'Souza D, Jayasinghe RG, Cheloni G, Bakhtiari M, Pabustan N, Nie K, Foltz JA, Saldarriaga I, Alaaeldin R, Lepisto E, Chen R, Fiala MA, Thomas BE, Cook A, Dos Santos JV, Chiang IL, Figueiredo I, Fortier J, Slade M, Oh ST, Rettig MP, Anderson E, Li Y, Dasari S, Strausbauch MA, Simon VA, Immune Atlas Consortium, Rahman AH, Chen Z, Lagana A, DiPersio JF, Rosenblatt J, Kim-Schulze S, Dhodapkar MV, Lonial S, Kumar S, Bhasin SS, Kourelis T, Vij R, Avigan D, Cho HJ, Mulligan G, Ding L, Gnjatic S, Vlachos IS, Bhasin M. A single-cell atlas characterizes dysregulation of the bone marrow immune microenvironment associated with outcomes in multiple myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.593193. [PMID: 38798338 PMCID: PMC11118283 DOI: 10.1101/2024.05.15.593193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Multiple Myeloma (MM) remains incurable despite advances in treatment options. Although tumor subtypes and specific DNA abnormalities are linked to worse prognosis, the impact of immune dysfunction on disease emergence and/or treatment sensitivity remains unclear. We established a harmonized consortium to generate an Immune Atlas of MM aimed at informing disease etiology, risk stratification, and potential therapeutic strategies. We generated a transcriptome profile of 1,149,344 single cells from the bone marrow of 263 newly diagnosed patients enrolled in the CoMMpass study and characterized immune and hematopoietic cell populations. Associating cell abundances and gene expression with disease progression revealed the presence of a proinflammatory immune senescence-associated secretory phenotype in rapidly progressing patients. Furthermore, signaling analyses suggested active intercellular communication involving APRIL-BCMA, potentially promoting tumor growth and survival. Finally, we demonstrate that integrating immune cell levels with genetic information can significantly improve patient stratification.
Collapse
Affiliation(s)
- William C. Pilcher
- Coultier Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Edgar Gonzalez-Kozlova
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yered Pita-Juarez
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dimitra Karagkouni
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Marina E Michaud
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | | | - Shivani Nanda
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yizhe Song
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Kazuhito Sato
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Julia T. Wang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Sarthak Satpathy
- Department of Biomedical Informatics, Emory School of Medicine, Atlanta, GA, USA
| | - Yuling Ma
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Darwin D'Souza
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Reyka G. Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Giulia Cheloni
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mojtaba Bakhtiari
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | | | - Kai Nie
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer A. Foltz
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Rania Alaaeldin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | | | - Rachel Chen
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark A. Fiala
- Bone Marrow Transplantation & Leukemia Section, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Beena E Thomas
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | | | - Junia Vieira Dos Santos
- Tisch Cancer Institute, Department of Immunology and Immunotherapy, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - I-ling Chiang
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Igor Figueiredo
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Fortier
- Bone Marrow Transplantation & Leukemia Section, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael Slade
- Bone Marrow Transplantation & Leukemia Section, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen T. Oh
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Immunomonitoring Laboratory, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael P. Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Ying Li
- Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Adeeb H Rahman
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhihong Chen
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessandro Lagana
- Tisch Cancer Institute, Department of Immunology and Immunotherapy, Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F. DiPersio
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jacalyn Rosenblatt
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center & Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madhav V Dhodapkar
- Department of Hematology Oncology, Emory School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute, Emory School of Medicine, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta
| | | | - Swati S Bhasin
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
| | | | - Ravi Vij
- Bone Marrow Transplantation & Leukemia Section, Division of Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - David Avigan
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center & Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO, USA
| | - Sacha Gnjatic
- Human Immune Monitoring Center, Tisch Cancer Institute, Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ioannis S Vlachos
- Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Spatial Technologies Unit, Harvard Medical School Initiative for RNA Medicine, Boston, MA, USA
- Cancer Center & Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA
| | - Manoj Bhasin
- Coultier Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA, USA
- Department of Biomedical Informatics, Emory School of Medicine, Atlanta, GA, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA, USA
| |
Collapse
|
38
|
Choi ME, Jung JM, Kim DH, Won CH, Chang SE, Lee MW, Lee WJ. Baseline Serum neutrophil-to-lymphocyte ratio in acral melanoma compared with nonacral melanoma and its prognostic significance. J Am Acad Dermatol 2024; 90:977-985. [PMID: 38272394 DOI: 10.1016/j.jaad.2023.12.064] [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: 09/24/2023] [Revised: 11/21/2023] [Accepted: 12/16/2023] [Indexed: 01/27/2024]
Abstract
BACKGROUND Acral lentiginous melanoma (ALM), a cutaneous melanoma subtype, exhibits a poorer prognosis than nonacral cutaneous melanoma (NACM). The neutrophil-to-lymphocyte ratio (NLR) is emerging as a prognostic indicator across diverse cancers. OBJECTIVE We explored the baseline NLR disparities between ALM and NACM, and the NLR's prognostic significance in patients with ALM. METHODS We reviewed records of patients with ALM and NACM diagnosed between 1997 and 2022, analyzing medical data. RESULTS Among 327 and 159 patients with ALM and NACM, respectively, baseline NLR varied based on distinct clinicopathologic factors between ALM and NACM. In stage 3 to 4 melanomas, the median NLR for ALM (2.18; IQR, 1.70-3.08) significantly surpassed NACM (1.74; IQR, 1.33-2.53) (P = .029). In patients with ALM, high NLR (hazard ratio, 1.64; 95% CI, 1.02-2.66; P = .043) was independently correlated with poor progression-free survival when adjusting for ulceration, Breslow thickness of ≥2 mm, and nodal invasion. LIMITATIONS Single-center, retrospective design. CONCLUSION Advanced-stage ALM exhibited a significantly higher baseline NLR compared with that of NACM. Evaluating baseline NLR could provide valuable prognostic insights for patients with ALM.
Collapse
Affiliation(s)
- Myoung Eun Choi
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Min Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Do Hyung Kim
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi Woo Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Woo Jin Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Michaelsen GL, da Silva LDRE, de Lima DS, Jaeger MDC, Brunetto AT, Dalmolin RJS, Sinigaglia M. A Prognostic Methylation-Driven Two-Gene Signature in Medulloblastoma. J Mol Neurosci 2024; 74:47. [PMID: 38662144 DOI: 10.1007/s12031-024-02203-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 02/21/2024] [Indexed: 04/26/2024]
Abstract
Medulloblastoma (MB) is one of the most common pediatric brain tumors and it is estimated that one-third of patients will not achieve long-term survival. Conventional prognostic parameters have limited and unreliable correlations with MB outcome, presenting a major challenge for patients' clinical improvement. Acknowledging this issue, our aim was to build a gene signature and evaluate its potential as a new prognostic model for patients with the disease. In this study, we used six datasets totaling 1679 samples including RNA gene expression and DNA methylation data from primary MB as well as control samples from healthy cerebellum. We identified methylation-driven genes (MDGs) in MB, genes whose expression is correlated with their methylation. We employed LASSO regression, incorporating the MDGs as a parameter to develop the prognostic model. Through this approach, we derived a two-gene signature (GS-2) of candidate prognostic biomarkers for MB (CEMIP and NCBP3). Using a risk score model, we confirmed the GS-2 impact on overall survival (OS) with Kaplan-Meier analysis. We evaluated its robustness and accuracy with receiver operating characteristic curves predicting OS at 1, 3, and 5 years in multiple independent datasets. The GS-2 showed highly significant results as an independent prognostic biomarker compared to traditional MB markers. The methylation-regulated GS-2 risk score model can effectively classify patients with MB into high and low-risk, reinforcing the importance of this epigenetic modification in the disease. Such genes stand out as promising prognostic biomarkers with potential application for MB treatment.
Collapse
Affiliation(s)
- Gustavo Lovatto Michaelsen
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - Lívia Dos Reis Edinger da Silva
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Federal University of Health Sciences of Porto Alegre, Porto Alegre, 90050-170, RS, Brazil
| | - Douglas Silva de Lima
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, 90035-003, RS, Brazil
| | - Mariane da Cunha Jaeger
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - André Tesainer Brunetto
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil
| | - Rodrigo Juliani Siqueira Dalmolin
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil
- Department of Biochemistry, Federal University of Rio Grande do Norte, Natal, 59064-741, RN, Brazil
| | - Marialva Sinigaglia
- Children's Cancer Institute, Porto Alegre, 90620-110, RS, Brazil.
- Bioinformatics Multidisciplinary Environment-BioME, Digital Metropole Institute, Federal University of Rio Grande do Norte, Natal, 59076-550, RN, Brazil.
- National Science and Technology Institute for Children's Cancer Biology and Pediatric Oncology - INCT BioOncoPed, Porto Alegre, 90035-003, RS, Brazil.
| |
Collapse
|
40
|
Martin D, Rödel F, Hehlgans S, Looso M, Ziegler PK, Fleischmann M, Diefenhardt M, Fries L, Kalinauskaite G, Tinhofer I, Zips D, Gani C, Rödel C, Fokas E. Inflammatory pathways confer resistance to chemoradiotherapy in anal squamous cell carcinoma. NPJ Precis Oncol 2024; 8:93. [PMID: 38653773 DOI: 10.1038/s41698-024-00585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/22/2024] [Indexed: 04/25/2024] Open
Abstract
Anal squamous cell carcinoma (ASCC) is associated with immunosuppression and infection with human papillomavirus (HPV). Response to standard chemoradiotherapy (CRT) varies considerably. A comprehensive molecular characterization of CRT resistance is lacking, and little is known about the interplay between tumor immune contexture, host immunity, and immunosuppressive and/or immune activating effects of CRT. Patients with localized ASCC, treated with CRT at three different sites of the German Cancer Consortium (DKTK) were included. Patient cohorts for molecular analysis included baseline formalin fixed paraffin embedded biopsies for immunohistochemistry (n = 130), baseline RNA sequencing (n = 98), peripheral blood immune profiling (n = 47), and serum cytokine measurement (n = 35). Gene set enrichment analysis showed that pathways for IFNγ, IFNα, inflammatory response, TNFα signaling via NF-κB, and EMT were significantly enriched in poor responders (all p < 0.001). Expression of interferon-induced transmembrane protein 1 (IFITM1), both on mRNA and protein levels, was associated with reduced Freedom from locoregional failure (FFLF, p = 0.037) and freedom from distant metastasis (FFDM, p = 0.014). An increase of PD-L1 expression on CD4+ T-cells (p < 0.001) and an increase in HLA-DR expression on T-cells (p < 0.001) was observed in the peripheral blood after CRT. Elevated levels of regulatory T-cells and CXCL2 were associated with reduced FFLF (p = 0.0044 and p = 0.004, respectively). Inflammatory pathways in tissue in line with elevated levels of regulatory T-cells and CXCL2 in peripheral blood are associated with resistance to CRT. To counteract this resistance mechanism, the RADIANCE randomized phase-2 trial currently tests the addition of the immune checkpoint inhibitor durvalumab to standard CRT in locally advanced ASCC.
Collapse
Affiliation(s)
- D Martin
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany.
| | - F Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - S Hehlgans
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - M Looso
- Max Planck Institute for Heart and Lung Research, Bioinformatics Core Unit, Bad Nauheim, Germany
| | - P K Ziegler
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- Dr. Senckenberg Institute of Pathology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - M Fleischmann
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - M Diefenhardt
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - L Fries
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - G Kalinauskaite
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, A Partnership between DKFZ and Charité University Hospital Berlin, Berlin, Germany
| | - I Tinhofer
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, A Partnership between DKFZ and Charité University Hospital Berlin, Berlin, Germany
| | - D Zips
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, A Partnership between DKFZ and Charité University Hospital Berlin, Berlin, Germany
| | - C Gani
- Eberhard Karls University, Tübingen, University Hospital Tübingen, Department of Radiation Oncology, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, A Partnership between DKFZ and University Hospital Tübingen, Tübingen, Germany
| | - C Rödel
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - E Fokas
- Department of Radiotherapy and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, A Partnership between DKFZ and University Hospital Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
41
|
Omar M, Xu Z, Rand SB, Alexanderani MK, Salles DC, Valencia I, Schaeffer EM, Robinson BD, Lotan TL, Loda M, Marchionni L. Semi-Supervised, Attention-Based Deep Learning for Predicting TMPRSS2:ERG Fusion Status in Prostate Cancer Using Whole Slide Images. Mol Cancer Res 2024; 22:347-359. [PMID: 38284821 PMCID: PMC10985477 DOI: 10.1158/1541-7786.mcr-23-0639] [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: 10/02/2023] [Revised: 12/26/2023] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
IMPLICATIONS Our study illuminates the potential of deep learning in effectively inferring key prostate cancer genetic alterations from the tissue morphology depicted in routinely available histology slides, offering a cost-effective method that could revolutionize diagnostic strategies in oncology.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Zhuoran Xu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sophie B. Rand
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Daniela C. Salles
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Itzel Valencia
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Tamara L. Lotan
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
42
|
Ramanovic M, Novak M, Perhavec A, Jordan T, Popuri K, Kozjek NR. Influence of nutritional status and body composition on postoperative events and outcome in patients treated for primary localized retroperitoneal sarcoma. Radiol Oncol 2024; 58:110-123. [PMID: 38378038 PMCID: PMC10878779 DOI: 10.2478/raon-2024-0013] [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: 07/26/2023] [Accepted: 11/03/2023] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Retroperitoneal sarcomas (RPS) are rare tumours of mesenchymal origin, commonly presented as a large tumour mass at time of diagnosis. We investigated the impact of body composition on outcome in patients operated on for primary localized RPS. PATIENTS AND METHODS We retrospectively analysed data for all patients operated on for primary RPS at our institution between 1999 and 2020. Preoperative skeletal muscle area (SMA), visceral and subcutaneous adipose tissue area (VAT and SAT) and muscle radiation attenuation (MRA) were calculated using computed tomography scans at the level of third lumbar vertebra. European Working Group on Sarcopenia in Older People (EWGSOP2) criteria were applied to define myopenia. Using maximum log-rank statistic method we determined the optimal cut-off values of body composition parameters. Myosteatosis was defined based on determined MRA cut-offs. RESULTS In total 58 patient were eligible for the study. With a median follow-up of 116 months, the estimated 5-year overall survival (OS) and local-recurrence free survival (LRFS) were 66.8% and 77.6%, respectively. Patients with myopenia had significantly lower 5-year OS compared to non-myopenic (p = 0.009). Skeletal muscle index and subcutaneous adipose tissue index predicted LRFS on univariate analysis (p = 0.052 and p = 0.039, respectively). In multivariate analysis high visceral-to-subcutaneous adipose tissue area ratio (VSR) independently predicted higher postoperative complication rate (89.2% vs. 10.8%, p = 0.008). Myosteatosis was associated with higher postoperative morbidity. CONCLUSIONS Myopenia affected survival, but not postoperative outcome in RPS. Visceral obesity, VSR (> 0.26) and myosteatosis were associated with higher postoperative morbidity. VSR was better prognostic factor than VAT in RPS.
Collapse
Affiliation(s)
- Manuel Ramanovic
- Biotechnical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Marko Novak
- Department of Surgical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Andraz Perhavec
- Department of Surgical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Taja Jordan
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Department for Radiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Karteek Popuri
- Department of Computer Science, Memorial University of Newfoundland, Newfundland, Canada
| | - Nada Rotovnik Kozjek
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Department of Clinical Nutrition, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
43
|
Varzaru B, Iacob RA, Bunduc S, Manea I, Sorop A, Spiridon A, Chelaru R, Croitoru A, Topala M, Becheanu G, Dumbrava M, Dima S, Popescu I, Gheorghe C. Prognostic Value of Circulating Cell-Free DNA Concentration and Neutrophil-to-Lymphocyte Ratio in Patients with Pancreatic Ductal Adenocarcinoma: A Prospective Cohort Study. Int J Mol Sci 2024; 25:2854. [PMID: 38474101 DOI: 10.3390/ijms25052854] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Circulating cell-free DNA (ccfDNA) quantity correlates with the clinical characteristics and prognosis of various cancer types. We investigated whether ccfDNA levels and the neutrophil-to-lymphocyte ratio (NLR) have prognostic value in patients with pancreatic ductal adenocarcinoma (PDAC). Peripheral blood was collected from 82 patients with PDAC prior to any diagnostic procedure or the administration of chemotherapy. Plasma DNA was isolated, and ccfDNA concentration and NLR were determined. We found that ccfDNA levels were correlated with age and tumor burden. Moreover, higher values of NLR (≥3.31) were linked with worse overall survival (OS) (4 vs. 10 months; log rank p = 0.011), and an elevated ccfDNA concentration (≥25.79 ng/mL) was strongly associated with shorter OS (4 vs. 8 months; log rank p = 0.009). According to the results of the multivariable Cox regression analysis, the baseline concentration of ccfDNA was an independent prognostic factor for OS (HR 0.45, 95% CI 0.21-0.97, p = 0.041). Furthermore, the combination of ccfDNA levels with NLR greatly enhanced the prognostic accuracy of PDAC patients. Our study demonstrates that ccfDNA concentration and NLR are independent predictors of survival in PDAC. Subsequent studies should validate this combination as a prognostic indicator in PDAC patients and assess its utility for guiding therapeutic decisions.
Collapse
Affiliation(s)
- Bianca Varzaru
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Razvan Andrei Iacob
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Stefania Bunduc
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Ioana Manea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Andrei Sorop
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Andreea Spiridon
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Raluca Chelaru
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Adina Croitoru
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Mihaela Topala
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Gabriel Becheanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Mona Dumbrava
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Simona Dima
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Irinel Popescu
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| | - Cristian Gheorghe
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Digestive Diseases and Liver Transplantation Center, Fundeni Clinical Institute, 022238 Bucharest, Romania
| |
Collapse
|
44
|
Wasilewski D, Onken J, Höricke P, Bukatz J, Murad S, Früh A, Shaked Z, Misch M, Kühl A, Klein O, Ehret F, Kaul D, Radbruch H, Capper D, Vajkoczy P, Horst D, Frost N, Bischoff P. Predictive role of intracranial PD-L1 expression in a real-world cohort of NSCLC patients treated with immune checkpoint inhibition following brain metastasis resection. J Neurooncol 2024; 167:155-167. [PMID: 38358406 PMCID: PMC10978684 DOI: 10.1007/s11060-024-04590-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 01/27/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Emerging evidence suggests that treatment of NSCLC brain metastases with immune checkpoint inhibitors (ICIs) is associated with response rates similar to those of extracranial disease. Programmed death-ligand 1 (PD-L1) tumor proportion score (TPS) serves as a predictive biomarker for ICI response. However, the predictive value of brain metastasis-specific (intracranial) PD-L1 TPS is not established. We investigated the role of intra- and extracranial PD-L1 TPS in NSCLC patients treated with ICI following brain metastasis resection. METHODS Clinical data from NSCLC patients treated with ICI following brain metastasis resection (n = 64) were analyzed. PD-L1 TPS of brain metastases (n = 64) and available matched extracranial tumor tissue (n = 44) were assessed via immunohistochemistry. Statistical analyses included cut point estimation via maximally selected rank statistics, Kaplan-Meier estimates, and multivariable Cox regression analysis for intracranial progression-free survival (icPFS), extracranial progression-free survival (ecPFS), and overall survival (OS). RESULTS PD-L1 expression was found in 54.7% of brain metastases and 68.2% of extracranial tumor tissues, with a median intra- and extracranial PD-L1 TPS of 7.5% (0 - 50%, IQR) and 15.0% (0 - 80%, IQR), respectively. In matched tissue samples, extracranial PD-L1 TPS was significantly higher than intracranial PD-L1 TPS (p = 0.013). Optimal cut points for intracranial and extracranial PD-L1 TPS varied according to outcome parameter assessed. Notably, patients with a high intracranial PD-L1 TPS (> 40%) exhibited significantly longer icPFS as compared to patients with a low intracranial PD-L1 TPS (≤ 40%). The cut point of 40% for intracranial PD-L1 TPS was independently associated with OS, icPFS and ecPFS in multivariable analyses. CONCLUSION Our study highlights the potential role of intracranial PD-L1 TPS in NSCLC, which could be used to predict ICI response in cases where extracranial tissue is not available for PD-L1 assessment as well as to specifically predict intracranial response.
Collapse
Affiliation(s)
- David Wasilewski
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany.
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Julia Onken
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Paul Höricke
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Jan Bukatz
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Selin Murad
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Anton Früh
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Zoe Shaked
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Martin Misch
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Anja Kühl
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Oliver Klein
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Felix Ehret
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - David Kaul
- Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Helena Radbruch
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - David Capper
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Neuropathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - David Horst
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Nikolaj Frost
- Charité Comprehensive Cancer Center, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
- Department of Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Philip Bischoff
- German Cancer Consortium (DKTK), Partner Site Berlin, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Berlin Institute of Health (BIH) Charité, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| |
Collapse
|
45
|
Pugh S, Fosdick BK, Nehring M, Gallichotte EN, VandeWoude S, Wilson A. Estimating cutoff values for diagnostic tests to achieve target specificity using extreme value theory. BMC Med Res Methodol 2024; 24:30. [PMID: 38331732 PMCID: PMC10851584 DOI: 10.1186/s12874-023-02139-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 12/28/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Rapidly developing tests for emerging diseases is critical for early disease monitoring. In the early stages of an epidemic, when low prevalences are expected, high specificity tests are desired to avoid numerous false positives. Selecting a cutoff to classify positive and negative test results that has the desired operating characteristics, such as specificity, is challenging for new tests because of limited validation data with known disease status. While there is ample statistical literature on estimating quantiles of a distribution, there is limited evidence on estimating extreme quantiles from limited validation data and the resulting test characteristics in the disease testing context. METHODS We propose using extreme value theory to select a cutoff with predetermined specificity by fitting a Pareto distribution to the upper tail of the negative controls. We compared this method to five previously proposed cutoff selection methods in a data analysis and simulation study. We analyzed COVID-19 enzyme linked immunosorbent assay antibody test results from long-term care facilities and skilled nursing staff in Colorado between May and December of 2020. RESULTS We found the extreme value approach had minimal bias when targeting a specificity of 0.995. Using the empirical quantile of the negative controls performed well when targeting a specificity of 0.95. The higher target specificity is preferred for overall test accuracy when prevalence is low, whereas the lower target specificity is preferred when prevalence is higher and resulted in less variable prevalence estimation. DISCUSSION While commonly used, the normal based methods showed considerable bias compared to the empirical and extreme value theory-based methods. CONCLUSIONS When determining disease testing cutoffs from small training data samples, we recommend using the extreme value based-methods when targeting a high specificity and the empirical quantile when targeting a lower specificity.
Collapse
Affiliation(s)
- Sierra Pugh
- Department of Statistics, Colorado State University, 102 Statistics Building, Fort Collins, 80523, Colorado, USA
| | - Bailey K Fosdick
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado, USA
| | - Mary Nehring
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Emily N Gallichotte
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Sue VandeWoude
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Ander Wilson
- Department of Statistics, Colorado State University, 102 Statistics Building, Fort Collins, 80523, Colorado, USA.
| |
Collapse
|
46
|
Shiomi K, Ichinoe M, Ushiwata A, Eshima K, Nagashio R, Hayashi S, Sonoda D, Kondo Y, Maruyama R, Mikubo M, Murakumo Y, Satoh Y. Insight into the significance of CD8+ tumor-infiltrating lymphocytes in squamous cell lung cancer. Thorac Cancer 2024; 15:299-306. [PMID: 38124453 PMCID: PMC10834194 DOI: 10.1111/1759-7714.15187] [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: 10/28/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Although there are great expectations regarding the use of tumor-infiltrating lymphocytes (TILs) to predict effects of immunotherapies and prognosis, knowledge about TILs remains insufficient for clinical application. METHODS We objectively investigated the prognostic significance of tumor-infiltrating CD8 + lymphocytes (CD8 + TILs) in squamous cell lung cancer (SQ-LC). Among patients who underwent surgical resection of SQ-LC in 2011-2017, 100 patients with pathological stage IA3-III were immunohistochemically studied to evaluate CD8 + TILs in the tumor stroma and parenchyma. The impact of CD8 + TILs on relapse-free survival was analyzed using a Kaplan-Meier survival analysis and multivariate analyses using Fine-Gray and Cox proportional hazards models. RESULTS The multivariate analysis showed that large and small numbers, but not intermediate numbers, of CD8 + TILs in the tumor stroma may be related to a more favorable prognosis (small vs. intermediate: HR, 0.64; 95% CI: 0.29-1.41, p = 0.27; large vs. intermediate: HR, 0.48; 95% CI: 0.21-1.09, p = 0.08). In contrast, a large number of CD8 + TILs in the tumor parenchyma was associated with a poor prognosis (HR, 2.60; 95% CI: 0.91-7.42, p = 0.075). An exploratory analysis showed a potentially strong association between an extremely large number of CD8 + TILs in the tumor parenchyma and a poor prognosis, even with a large number of CD8 + TILs in the tumor stroma. CONCLUSION Our study provided partial but important information on the significance of CD8 + TILs in SQ-LC. To use CD8 + TILs as biomarkers, a better understanding of CD8 + TILs as well as other important components in the tumor microenvironment and the inflammatory phenotypes they form may be needed.
Collapse
Affiliation(s)
- Kazu Shiomi
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Masaaki Ichinoe
- Department of PathologyKitasato University School of MedicineSagamihara‐shiJapan
| | - Ai Ushiwata
- Department of Clinical Medicine (Biostatistics)Kitasato University School of PharmacyTokyoJapan
| | - Koji Eshima
- Department of BiosciencesKitasato University School of SciencesSagamihara‐shiJapan
| | - Ryo Nagashio
- Department of Applied Tumor Pathology, Graduate School of Medical SciencesKitasato UniversitySagamihara‐shiJapan
| | - Shoko Hayashi
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Dai Sonoda
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Yasuto Kondo
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Raito Maruyama
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Masashi Mikubo
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| | - Yoshiki Murakumo
- Department of PathologyKitasato University School of MedicineSagamihara‐shiJapan
| | - Yukitoshi Satoh
- Department of Thoracic SurgeryKitasato University School of MedicineSagamihara‐shiJapan
| |
Collapse
|
47
|
Othman J, Tiong IS, O'Nions J, Dennis M, Mokretar K, Ivey A, Austin M, Latif AL, Amer M, Chan WY, Crawley C, Crolla F, Cross J, Dang R, Elliot J, Fong CY, Galli S, Gallipoli P, Hogan F, Kalkur P, Khan A, Krishnamurthy P, Laurie J, Loo S, Marshall S, Mehta P, Murthy V, Nagumantry S, Pillai S, Potter N, Sellar R, Taylor T, Zhao R, Russell NH, Wei AH, Dillon R. Molecular MRD is strongly prognostic in patients with NPM1-mutated AML receiving venetoclax-based nonintensive therapy. Blood 2024; 143:336-341. [PMID: 37647641 DOI: 10.1182/blood.2023021579] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/27/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023] Open
Abstract
ABSTRACT Assessment of measurable residual disease (MRD) by quantitative reverse transcription polymerase chain reaction is strongly prognostic in patients with NPM1-mutated acute myeloid leukemia (AML) treated with intensive chemotherapy; however, there are no data regarding its utility in venetoclax-based nonintensive therapy, despite high efficacy in this genotype. We analyzed the prognostic impact of NPM1 MRD in an international real-world cohort of 76 previously untreated patients with NPM1-mutated AML who achieved complete remission (CR)/CR with incomplete hematological recovery following treatment with venetoclax and hypomethylating agents (HMAs) or low-dose cytarabine (LDAC). A total of 44 patients (58%) achieved bone marrow (BM) MRD negativity, and a further 14 (18%) achieved a reduction of ≥4 log10 from baseline as their best response, with no difference between HMAs and LDAC. The cumulative rates of BM MRD negativity by the end of cycles 2, 4, and 6 were 25%, 47%, and 50%, respectively. Patients achieving BM MRD negativity by the end of cycle 4 had 2-year overall of 84% compared with 46% if MRD was positive. On multivariable analyses, MRD negativity was the strongest prognostic factor. A total of 22 patients electively stopped therapy in BM MRD-negative remission after a median of 8 cycles, with 2-year treatment-free remission of 88%. In patients with NPM1-mutated AML attaining remission with venetoclax combination therapies, NPM1 MRD provides valuable prognostic information.
Collapse
Affiliation(s)
- Jad Othman
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Ing S Tiong
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Alfred Hospital and Monash University, Melbourne, Australia
- Austin Health and Olivia Newton John Cancer Research Institute, Melbourne, Australia
| | - Jenny O'Nions
- Department of Haematology, University College London Hospital NHS Foundation Trust, London, United Kingdom
| | - Mike Dennis
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | | | - Adam Ivey
- Alfred Hospital and Monash University, Melbourne, Australia
| | - Michael Austin
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Anne-Louise Latif
- Department of Haematology, Queen Elizabeth University Hospital, Glasgow, United Kingdom
| | - Mariam Amer
- Haematology, University Hospital Southampton, Southampton, United Kingdom
| | - Wei Yee Chan
- Department of Haematology, University College London Hospital NHS Foundation Trust, London, United Kingdom
| | - Charles Crawley
- Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | | | - Joe Cross
- Haematology Department, University Hospital Bristol, Bristol, United Kingdom
| | - Ray Dang
- James Cook University Hospital, Middlesbrough, United Kingdom
| | | | - Chun Y Fong
- Austin Health and Olivia Newton John Cancer Research Institute, Melbourne, Australia
| | - Sofia Galli
- Frimley Park Hospital, London, United Kingdom
| | - Paolo Gallipoli
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | | | - Anjum Khan
- Department of Haematology, Leeds Teaching Hospitals Trust, Leeds, United Kingdom
| | | | | | - Sun Loo
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Scott Marshall
- City Hospitals Sunderland NHS Trust, Sunderland, United Kingdom
| | - Priyanka Mehta
- Haematology Department, University Hospital Bristol, Bristol, United Kingdom
| | - Vidhya Murthy
- Centre for Clinical Haematology, University Hospitals Birmingham, Birmingham, United Kingdom
| | | | - Srinivas Pillai
- Royal Stoke University Hospital, University Hospital of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | - Nicola Potter
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - Rob Sellar
- Department of Haematology, University College London Hospital NHS Foundation Trust, London, United Kingdom
| | - Tom Taylor
- Nottingham University Hospital, Nottingham, United Kingdom
| | - Rui Zhao
- Torbay Hospital, Torquay, United Kingdom
| | - Nigel H Russell
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | - Andrew H Wei
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Richard Dillon
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
- Department of Haematology, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
48
|
Penack O, Luft T, Peczynski C, Benner A, Sica S, Arat M, Itäla-Remes M, Corral LL, Schaap NPM, Karas M, Raida L, Schroeder T, Dreger P, Metafuni E, Ozcelik T, Sandmaier BM, Kordelas L, Moiseev I, Schoemans H, Koenecke C, Basak GW, Peric Z. Endothelial Activation and Stress Index (EASIX) to predict mortality after allogeneic stem cell transplantation: a prospective study. J Immunother Cancer 2024; 12:e007635. [PMID: 38199608 PMCID: PMC10806535 DOI: 10.1136/jitc-2023-007635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND We previously reported that the "Endothelial Activation and Stress Index" (EASIX; ((creatinine×lactate dehydrogenase)÷thrombocytes)) measured before start of conditioning predicts mortality after allogeneic hematopoietic stem cell transplantation (alloSCT) when used as continuous score. For broad clinical implementation, a prospectively validated EASIX-pre cut-off is needed that defines a high-risk cohort and is easy to use. METHOD In the current study, we first performed a retrospective cohort analysis in n=2022 alloSCT recipients and identified an optimal cut-off for predicting non-relapse mortality (NRM) as EASIX-pre=3. For cut-off validation, we conducted a multicenter prospective study with inclusion of n=317 first alloSCTs from peripheral blood stem cell in adult patients with acute leukemia, lymphoma or myelodysplastic syndrome/myeloproliferative neoplasms in the European Society for Blood and Marrow Transplantation network. RESULTS Twenty-three % (n=74) of alloSCT recipients had EASIX-pre ≥3 taken before conditioning. NRM at 2 years was 31.1% in the high EASIX group versus 11.5% in the low EASIX group (p<0.001). Patients with high EASIX-pre also had worse 2 years overall survival (51.6% vs 70.9%; p=0.002). We were able to validate the cut-off and found that EASIX ≥3 was associated with more than twofold increased risk for NRM in multivariate analysis (HR=2.18, 95% CI 1.2 to 3.94; p=0.01). No statistically significant difference could be observed for the incidence of relapse. CONCLUSIONS The results of this study provide a prospectively validated standard laboratory biomarker index to estimate the transplant-related mortality risk after alloSCT. EASIX ≥3 taken before conditioning identifies a population of alloSCT recipients who have a more than twofold increased risk of treatment-related mortality.
Collapse
Affiliation(s)
- Olaf Penack
- Department for Haematology, Oncology and Tumorimmunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- EBMT Transplant Complications Working Party, Heidelberg, Germany
| | - Thomas Luft
- Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | - Christophe Peczynski
- EBMT Transplant Complications Working Party, Paris, France
- Department of Haematology, Sorbonne University, Paris, France
| | - Axel Benner
- German Cancer Research Centre, Heidelberg, Germany
| | - Simona Sica
- Istituto di Ematologia, Universita Cattolica S. Cuore, Rome, Italy
| | - Mutlu Arat
- Florence Nightingale Hospital, Hematopoietic SCT Unit, Demiroglu Bilim University Istanbul, Istanbul, Turkey
| | | | - Lucia López Corral
- Department for Haematology, Hospital Clinico San Carlos, Salamanca, Spain
| | | | - Michal Karas
- Hospital Dept. of Hematology/Oncology, Charles University, Pilsen, Czech Republic
| | - Ludek Raida
- Olomouc University Social Health Institute, Olomouc, Czech Republic
| | - Thomas Schroeder
- Dept. of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Peter Dreger
- Medicine V, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Tulay Ozcelik
- Florence Nightingale Hospital, Hematopoietic SCT Unit, Demiroglu Bilim University Istanbul, Istanbul, Turkey
| | | | - Lambros Kordelas
- Dept. of Hematology and Stem Cell Transplantation, University Hospital Essen, Essen, Germany
| | - Ivan Moiseev
- EBMT Transplant Complications Working Party, Paris, France
- First Pavlov State Medical University of St Petersburg, St Petersburg, Russian Federation
| | - Hélène Schoemans
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, University Hospitals Leuven and KU Leuven, Leuven, Belgium
| | - Christian Koenecke
- Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Grzegorz W Basak
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, Oncology and Internal Medicine, the Medical University of Warsaw, Warsaw, Poland
| | - Zinaida Peric
- EBMT Transplant Complications Working Party, Paris, France
- Department of Hematology, University of Rijeka, Rijeka, Croatia
| |
Collapse
|
49
|
Huo RR, Pan LX, Wu PS, Liang XM, You XM, Ma L, Zhong JH. Prognostic value of aspartate aminotransferase/alanine aminotransferase ratio in hepatocellular carcinoma after hepatectomy. BJS Open 2024; 8:zrad155. [PMID: 38242573 PMCID: PMC10798825 DOI: 10.1093/bjsopen/zrad155] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/30/2023] [Accepted: 11/19/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND The prognostic significance of the aspartate aminotransferase/alanine aminotransferase (AST/ALT) ratio in hepatocellular carcinoma remains uncertain. The aim of the current study was to evaluate the association between the AST/ALT ratio and prognosis in patients with hepatocellular carcinoma after hepatectomy, and to explore the role of underlying liver diseases as mediators. METHODS This retrospective study included patients with hepatocellular carcinoma who underwent hepatectomy between January 2014 and January 2018 at two Chinese hospitals. The maximally selected rank statistic and g-computation approach were used to quantify and visualize the association between the AST/ALT ratio and overall survival or recurrence-free survival. The role of mediators (chronic hepatitis B, hepatic steatosis and liver cirrhosis) was analysed. RESULTS Among the 1519 patients (mean(s.d.) age at baseline, 50.5(11.3) years), 1309 (86.2%) were male. During a median follow-up of 46.0 months, 514 (33.8%) patients died and 358 (23.6%) patients experienced recurrence. The optimal cut-off value for the AST/ALT ratio was 1.4, and the AST/ALT ratio greater than or equal to 1.4 was independently associated with a 39.0% increased risk of death and a 30.0% increased risk of recurrence (overall survival: hazard ratio (HR), 1.39; 95% c.i. 1.15 to 1.68; recurrence-free survival: HR, 1.30; 95% c.i. 1.12 to 1.52) after adjusting for confounders. Chronic hepatitis B significantly mediated the association of the ratio of AST/ALT with both overall survival and recurrence-free survival (20.3% for overall survival; 20.1% for recurrence-free survival). CONCLUSION The AST/ALT ratio greater than or equal to 1.4 was associated with shorter overall survival and recurrence-free survival in patients with hepatocellular carcinoma after hepatectomy, and chronic hepatitis B may play a role in their association.
Collapse
Affiliation(s)
- Rong-Rui Huo
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Li-Xin Pan
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Pei-Sheng Wu
- Hepatobiliary Surgery Department, The First People’s Hospital of Qinzhou, Qinzhou, China
| | - Xiu-Mei Liang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Xue-Mei You
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour (Guangxi Medical University), Nanning, China
| | - Liang Ma
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jian-Hong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumour (Guangxi Medical University), Nanning, China
| |
Collapse
|
50
|
Yang H, Qiu W, Liu Z. Anoikis-related mRNA-lncRNA and DNA methylation profiles for overall survival prediction in breast cancer patients. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:1590-1609. [PMID: 38303479 DOI: 10.3934/mbe.2024069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
As a type of programmed cell death, anoikis resistance plays an essential role in tumor metastasis, allowing cancer cells to survive in the systemic circulation and as a key pathway for regulating critical biological processes. We conducted an exploratory analysis to improve risk stratification and optimize adjuvant treatment choices for patients with breast cancer, and identify multigene features in mRNA and lncRNA transcriptome profiles associated with anoikis. First, the variance selection method filters low information content genes in RNA sequence and then extracts the mRNA and lncRNA expression data base on annotation files. Then, the top ten key mRNAs are screened out through the PPI network. Pearson analysis has been employed to identify lncRNAs related to anoikis, and the prognosis-related lncRNAs are selected using Univariate Cox regression and machine learning. Finally, we identified a group of RNAs (including ten mRNAs and six lncRNAs) and integrated the expression data of 16 genes to construct a risk-scoring system for BRCA prognosis and drug sensitivity analysis. The risk score's validity has been evaluated with the ROC curve, Kaplan-Meier survival curve analysis and decision curve analysis (DCA). For the methylation data, we have obtained 169 anoikis-related prognostic methylation sites, integrated these sites with 16 RNA features and further used the deep learning model to evaluate and predict the survival risk of patients. The developed anoikis feature is demonstrated a consistency index (C-index) of 0.778, indicating its potential to predict the survival probability of breast cancer patients using deep learning methods.
Collapse
Affiliation(s)
- Huili Yang
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China
| | - Wangren Qiu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China
| | - Zi Liu
- Computer Department, Jingdezhen Ceramic University, Jingdezhen 333403, China
| |
Collapse
|