1
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [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: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
2
|
Nencini F, Giurranna E, Borghi S, Taddei N, Fiorillo C, Becatti M. Fibrinogen Oxidation and Thrombosis: Shaping Structure and Function. Antioxidants (Basel) 2025; 14:390. [PMID: 40298646 PMCID: PMC12024030 DOI: 10.3390/antiox14040390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/19/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Fibrinogen, a pivotal plasma glycoprotein, plays an essential role in hemostasis by serving as the precursor to fibrin, which forms the structural framework of blood clots. Beyond coagulation, fibrinogen influences immune responses, inflammation, and tissue repair. Oxidative stress, characterized by an imbalance between reactive oxygen species (ROS) and antioxidants, induces fibrinogen oxidation, significantly altering its structure and function. This narrative review synthesizes findings from in vitro, ex vivo, and clinical studies, emphasizing the impact of fibrinogen oxidation on clot formation, architecture, and degradation. Oxidative modifications result in denser fibrin clots with thinner fibers, reduced permeability, and heightened resistance to fibrinolysis. These structural changes exacerbate prothrombotic conditions in cardiovascular diseases, diabetes, chronic inflammatory disorders and cancer. In contrast, "low-dose" oxidative stress may elicit protective adaptations in fibrinogen, preserving its function. The review also highlights discrepancies in experimental findings due to variability in oxidation protocols and patient conditions. Understanding the interplay between oxidation and fibrinogen function could unveil therapeutic strategies targeting oxidative stress. Antioxidant therapies or selective inhibitors of detrimental oxidation hold potential for mitigating thrombotic risks. However, further research is essential to pinpoint specific fibrinogen oxidation sites, clarify their roles in clot dynamics, and bridge the gap between basic research and clinical practice.
Collapse
|
3
|
Șerban RE, Popescu DM, Boldeanu MV, Florescu DN, Șerbănescu MS, Șandru V, Panaitescu-Damian A, Forțofoiu D, Șerban RC, Gherghina FL, Vere CC. The Diagnostic and Prognostic Role of Inflammatory Markers, Including the New Cumulative Inflammatory Index (IIC) and Mean Corpuscular Volume/Lymphocyte (MCVL), in Colorectal Adenocarcinoma. Cancers (Basel) 2025; 17:990. [PMID: 40149324 PMCID: PMC11940412 DOI: 10.3390/cancers17060990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Colorectal cancer affects a large number of patients worldwide, with numerous factors being involved in its etiopathogenesis and chronic inflammation playing an essential role in tumor development. In this study, we analyzed and compared several markers of inflammation that are relatively easy to obtain for a rapid and accurate diagnosis and prognosis. Methods: This study included 219 patients diagnosed with colorectal cancer, analyzing the inflammation scores derived from their blood cells and inflammatory circulating proteins. These inflammatory markers are neutrophil-to-lymphocyte ratio-NLR; platelet-to-lymphocyte ratio-PLR; lymphocyte-to-monocyte ratio-LMR; systemic immune inflammation index-SII; systemic inflammatory response index-SIRI; aggregate index of systemic inflammation-AISI; derived neutrophil-to-lymphocyte ratio-dNLR; C-reactive protein-to-albumin ratio-CAR; and fibrinogen-to-albumin ratio-FAR. In the analysis of patients with colorectal cancer, we have also introduced two new recently developed inflammatory markers: the cumulative inflammatory index (IIC) and the ratio between the mean corpuscular volume and lymphocytes (MCVL). This study aimed to correlate the inflammatory markers' levels with the colorectal cancer diagnostic stage, the tumor and clinical characteristics of the colorectal cancer patients, and 36 months' survival time and to evaluate the diagnostic and prognostic capacity and accuracy of these inflammatory markers in this type of cancer. Results: We showed that the levels of the analyzed inflammation markers correlate with the TNM stage, the tumor pathological differentiation grade, the age and gender of the patients, and overall survival, with their increased levels being associated with a lower survival rate. Conclusions: The analyzed markers, which are easy to perform right from the patient's admission, can be helpful both in diagnosis and, mostly, in prognosis, sustaining the role of inflammation in cancer. By comparing them, we showed which one can be useful for increased sensitivity and specificity in the diagnosis and prognosis of colorectal cancer patients.
Collapse
Affiliation(s)
- Robert-Emmanuel Șerban
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.-E.Ș.); (D.N.F.); (C.-C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dragoș-Marian Popescu
- Department of Extreme Conditions Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihail-Virgil Boldeanu
- Department of Immunology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dan Nicolae Florescu
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.-E.Ș.); (D.N.F.); (C.-C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
| | - Mircea-Sebastian Șerbănescu
- Department of Medical Informatics and Biostatistics, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Vasile Șandru
- Department of Gastroenterology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Clinical Department of Gastroenterology, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| | - Afrodita Panaitescu-Damian
- Clinical Department of Gastroenterology, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| | - Dragoș Forțofoiu
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Rebecca-Cristiana Șerban
- Department of Cellular and Molecular Biology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Florin-Liviu Gherghina
- Department of Medical Rehabilitation, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Cristin-Constantin Vere
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (R.-E.Ș.); (D.N.F.); (C.-C.V.)
- Research Center of Gastroenterology and Hepatology, University of Medicine and Pharmacy of Craiova, 200638 Craiova, Romania
| |
Collapse
|
4
|
Zhang Y, Li Z, Zhang J, Mafa T, Zhang J, Zhu H, Chen L, Zong Z, Yang L. Fibrinogen: A new player and target on the formation of pre-metastatic niche in tumor metastasis. Crit Rev Oncol Hematol 2025; 207:104625. [PMID: 39826884 DOI: 10.1016/j.critrevonc.2025.104625] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
Tumor metastasis involves a series of complex and coordinated processes, which is the main cause of patient death and still a significant challenge in cancer treatment. Pre-metastatic niches (PMN), a specialized microenvironment that develops in distant organs prior to the arrival of metastatic cancer cells, plays a crucial role in driving tumor metastasis. The development of PMN depends on a complex series of cellular and molecular components including tumor-derived factors, bone marrow-derived cells, resident immune cells, and extracellular matrix. Fibrinogen, a key factor in the typical blood clotting process, is related to tumor metastasis and prognosis, according to a growing body of evidence in recent years. Fibrinogen has emerged as an important factor in mediating the formation of tumor microenvironment. Nevertheless, a clear and detailed mechanism by which fibrinogen promotes tumor metastasis remains unknown. In this review, we first explore the roles of fibrinogen in the development of PMN from four perspectives: immunosuppression, inflammation, angiogenesis, and extracellular matrix remodeling. We highlight the significance of fibrinogen in shaping PMN and discuss its potential therapeutic values, opening new avenues for targeting fibrinogen to prevent or treat metastasis.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Zelin Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The First Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Jiamao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Tatenda Mafa
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Jingyu Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang, Jiangxi 330006, China
| | - Hui Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China
| | - Lifang Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang, Jiangxi 330006, China
| | - Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
5
|
Kuras M, Betancourt LH, Hong R, Szadai L, Rodriguez J, Horvatovich P, Pla I, Eriksson J, Szeitz B, Deszcz B, Welinder C, Sugihara Y, Ekedahl H, Baldetorp B, Ingvar C, Lundgren L, Lindberg H, Oskolas H, Horvath Z, Rezeli M, Gil J, Appelqvist R, Kemény LV, Malm J, Sanchez A, Szasz AM, Pawłowski K, Wieslander E, Fenyö D, Nemeth IB, Marko-Varga G. Proteogenomic Profiling of Treatment-Naïve Metastatic Malignant Melanoma. Cancers (Basel) 2025; 17:832. [PMID: 40075679 PMCID: PMC11899103 DOI: 10.3390/cancers17050832] [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: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Melanoma is a highly heterogeneous disease, and a deeper molecular classification is essential for improving patient stratification and treatment approaches. Here, we describe the histopathology-driven proteogenomic landscape of 142 treatment-naïve metastatic melanoma samples to uncover molecular subtypes and clinically relevant biomarkers. METHODS We performed an integrative proteogenomic analysis to identify proteomic subtypes, assess the impact of BRAF V600 mutations, and study the molecular profiles and cellular composition of the tumor microenvironment. Clinical and histopathological data were used to support findings related to tissue morphology, disease progression, and patient outcomes. RESULTS Our analysis revealed five distinct proteomic subtypes that integrate immune and stromal microenvironment components and correlate with clinical and histopathological parameters. We demonstrated that BRAF V600-mutated melanomas exhibit biological heterogeneity, where an oncogene-induced senescence-like phenotype is associated with improved survival. This led to a proposed mortality risk-based stratification that may contribute to more personalized treatment strategies. Furthermore, tumor microenvironment composition strongly correlated with disease progression and patient outcomes, highlighting a histopathological connective tissue-to-tumor ratio assessment as a potential decision-making tool. We identified a melanoma-associated SAAV signature linked to extracellular matrix remodeling and SAAV-derived neoantigens as potential targets for anti-tumor immune responses. CONCLUSIONS This study provides a comprehensive stratification of metastatic melanoma, integrating proteogenomic insights with histopathological features. The findings may aid in the development of tailored diagnostic and therapeutic strategies, improving patient management and outcomes.
Collapse
Affiliation(s)
- Magdalena Kuras
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Lazaro Hiram Betancourt
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; (R.H.); (D.F.)
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Leticia Szadai
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.S.); (I.B.N.)
| | - Jimmy Rodriguez
- Department of Biochemistry and Biophysics, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - Peter Horvatovich
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
- Department of Analytical Biochemistry, Faculty of Science and Engineering, University of Groningen, 9712 CP Groningen, The Netherlands
| | - Indira Pla
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Jonatan Eriksson
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1085 Budapest, Hungary
| | - Bartłomiej Deszcz
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
| | - Charlotte Welinder
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Yutaka Sugihara
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Henrik Ekedahl
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
- SUS University Hospital Lund, 222 42 Lund, Sweden;
| | - Bo Baldetorp
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Christian Ingvar
- SUS University Hospital Lund, 222 42 Lund, Sweden;
- Department of Surgery, Clinical Sciences, Lund University, SUS, 221 00 Lund, Sweden
| | - Lotta Lundgren
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
- SUS University Hospital Lund, 222 42 Lund, Sweden;
| | - Henrik Lindberg
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Henriett Oskolas
- Department of Clinical Sciences Lund, Division of Oncology, Lund University, 221 00 Lund, Sweden; (C.W.); (B.B.); (L.L.); (H.O.)
| | - Zsolt Horvath
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Jeovanis Gil
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - Roger Appelqvist
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
| | - Lajos V. Kemény
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, 1085 Budapest, Hungary;
- Department of Dermatology, Venereology and Dermatooncology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- Department of Physiology, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
- MTA-SE Lendület “Momentum” Dermatology Research Group, Hungarian Academy of Sciences and Semmelweis University, 1085 Budapest, Hungary
| | - Johan Malm
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - Aniel Sanchez
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | | | - Krzysztof Pawłowski
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland;
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elisabet Wieslander
- Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, 214 28 Malmö, Sweden; (M.K.); (J.G.); (J.M.); (A.S.); (K.P.)
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY 10016, USA; (R.H.); (D.F.)
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - Istvan Balazs Nemeth
- Department of Dermatology and Allergology, University of Szeged, 6720 Szeged, Hungary; (L.S.); (I.B.N.)
| | - György Marko-Varga
- Department of Biomedical Engineering, Lund University, 221 00 Lund, Sweden; (P.H.); (I.P.); (J.E.); (Y.S.); (H.L.); (M.R.); (R.A.); (G.M.-V.)
- Chemical Genomics Global Research Lab, Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- 1st Department of Surgery, Tokyo Medical University, Tokyo 160-8402, Japan
| |
Collapse
|
6
|
Morales-Pacheco M, Valenzuela-Mayen M, Gonzalez-Alatriste AM, Mendoza-Almanza G, Cortés-Ramírez SA, Losada-García A, Rodríguez-Martínez G, González-Ramírez I, Maldonado-Lagunas V, Vazquez-Santillan K, González-Covarrubias V, Pérez-Plasencia C, Rodríguez-Dorantes M. The role of platelets in cancer: from their influence on tumor progression to their potential use in liquid biopsy. Biomark Res 2025; 13:27. [PMID: 39934930 DOI: 10.1186/s40364-025-00742-w] [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: 10/11/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Platelets, anucleate blood cells essential for hemostasis, are increasingly recognized for their role in cancer, challenging the traditional notion of their sole involvement in blood coagulation. It has been demonstrated that platelets establish bidirectional communication with tumor cells, contributing to tumor progression and metastasis through diverse molecular mechanisms such as modulation of proliferation, angiogenesis, epithelial-mesenchymal transition, resistance to anoikis, immune evasion, extravasation, chemoresistance, among other processes. Reciprocally, cancer significantly alters platelets in their count and composition, including mRNA, non-coding RNA, proteins, and lipids, product of both internal synthesis and the uptake of tumor-derived molecules. This phenomenon gives rise to tumor-educated platelets (TEPs), which are emerging as promising tools for the development of liquid biopsies. In this review, we provide a detailed overview of the dynamic roles of platelets in tumor development and progression as well as their use in diagnosis and prognosis. We also provide our view on current limitations, challenges and future research areas, including the need to design more efficient strategies for their isolation and analysis, as well as the validation of their sensitivity and specificity through large-scale and rigorous clinical trials. This research will not only enable the evaluation of their clinical viability but could also open new opportunities to enhance diagnostic accuracy and develop personalized treatments in oncology.
Collapse
Affiliation(s)
- Miguel Morales-Pacheco
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | - Miguel Valenzuela-Mayen
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | | | - Gretel Mendoza-Almanza
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Sergio A Cortés-Ramírez
- Department of Pharmacology and Toxicology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Alberto Losada-García
- Department of Pharmacology and Toxicology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Griselda Rodríguez-Martínez
- Laboratorio de Oncogenómica, Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
- Laboratorio de Investigación en Patógenos Respiratorios y Producción de Biológicos, Hospital Infantil de México Federico Gómez, Mexico City, 14610, Mexico
| | - Imelda González-Ramírez
- Departamento de Atención a La Salud, Universidad Autónoma Metropolitana Xochimilco, Mexico City, 14610, Mexico
| | - Vilma Maldonado-Lagunas
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Karla Vazquez-Santillan
- Laboratorio de Innovación en Medicina de Precisión, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Vanessa González-Covarrubias
- Laboratorio de Farmacogenómica, Instituto Nacional de Medicina Genómica, Secretaría de Salud, Mexico City, 14610, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica, FES-Iztacala, Universidad Nacional Autónoma de México (UNAM), Iztacala, Tlalnepantla, 54090, Mexico
| | | |
Collapse
|
7
|
Yu H, Ben-Akiva E, Meyer RA, Green JJ. Biomimetic Anisotropic-Functionalized Platelet-Membrane-Coated Polymeric Particles for Targeted Drug Delivery to Human Breast Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2025; 17:351-362. [PMID: 39713866 DOI: 10.1021/acsami.4c15471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Biomimetic particles that can replicate aspects of natural biological cell function are useful for advanced biological engineering applications. Engineering such particles requires mimicking the chemical complexity of the surface of biological cells, and this can be achieved by coating synthetic particles with naturally derived cell membranes. Past research has demonstrated the feasibility of utilizing cell membrane coatings from a variety of cell types to achieve extended blood circulation half-life. A particle's shape can also be designed to mimic a biological cell or virus, and this physical attribute can cause particular transport and biodistribution properties. However, the potential synergy between engineering a biomimetic particle's core shape in combination with functionalizing its surface with cell membranes to achieve targeted drug delivery has not been well-investigated. Here, anisotropic poly(lactic-co-glycolic acid) (PLGA) particles are coated with platelet membranes to engineer particles with enhanced stealth properties that are biomimetic in size, shape, and surface composition to natural platelets. The natural ability of platelets to target tumor cells was harnessed to develop a particulate system for targeted dual delivery of a small molecule and protein to cancer cells. The particles had targeted binding to metastatic human breast cancer cells, leading to enhanced killing of these cells in a mouse model through codelivery of TRAIL and doxorubicin. This system can be used for cancer cell killing and could potentially be utilized in preventing breast cancer metastasis. By engineering both the physical and chemical properties of the particles, biomimicry and therapeutic promise can be best achieved.
Collapse
Affiliation(s)
- Hongzhe Yu
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Elana Ben-Akiva
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Randall A Meyer
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, Johns Hopkins Translational ImmunoEngineering Center, and the Institute for Nanobiotechnology, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
- Departments of Oncology, Ophthalmology, and Neurosurgery, Bloomberg-Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
- Departments of Materials Science and Engineering and Chemical and Biomolecular Engineering, Johns Hopkins University, 400 N. Broadway, Smith Building 5017, Baltimore, Maryland 21231, United States
| |
Collapse
|
8
|
Monegatti S, Martinelli N, Friso S, Spronk HMH, Cate HT. Mechanisms and management of thrombosis in cancer: Focus on gastrointestinal malignancies. J Pharmacol Exp Ther 2025; 392:100018. [PMID: 39893001 DOI: 10.1124/jpet.124.002203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/14/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024] Open
Abstract
Cancer patients have an increased risk of venous thromboembolism, which is their second cause of death after disease progression itself. Several thrombotic risk factors coexist in cancer patients, including the ability of both cancer and tumoral microenvironment's cells to directly or indirectly activate platelets and the enzymes of the coagulation cascade, resulting in a hypercoagulable state of blood. This narrative review gives an overview of the main mechanisms leading to venous thromboembolism in cancer patients, including the role that platelets and the clotting proteins may have in tumor growth and metastasis. Of note, the hemostatic balance is altered in cancer patients who may, next to a thrombosis tendency, also have an increased risk of bleeding. To highlight the complexity and the precariousness of the hemostatic balance of these patients, we discuss 2 specific gastrointestinal malignancies: hepatocellular carcinoma, which is frequently associated with liver cirrhosis, a condition that causes profound alterations of hemostasis, and colorectal cancer, which is characterized by a fragile mucosa that is prone to bleeding. Understanding the molecular mechanisms of cancer-associated thrombosis may give a unique opportunity to develop new innovative drugs, acting differently on distinct pathways and potentially allowing to reduce the risk of bleeding related to antithrombotic therapies. SIGNIFICANCE STATEMENT: The topic is significant because understanding the molecular mechanisms leading to cancer-associated thrombosis and bleeding, focusing on gastrointestinal malignancies, enables the development of more rationale and innovative antithrombotic strategies for cancer-associated thrombosis. Eventually, this will support an improved and patient-tailored antithrombotic management in vulnerable oncologic patients.
Collapse
Affiliation(s)
- Simone Monegatti
- Department of Medicine, University of Verona, Verona, Italy; Departments of Internal Medicine and Biochemistry, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | | | | | - Henri M H Spronk
- Departments of Internal Medicine and Biochemistry, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands
| | - Hugo Ten Cate
- Departments of Internal Medicine and Biochemistry, Maastricht University Medical Centre and CARIM School for Cardiovascular Diseases, Maastricht, the Netherlands.
| |
Collapse
|
9
|
Kottana RK, Schnoor B, Papa A. A method to quantitatively characterize the formation and dissociation of tumor cell clusters using light transmission aggregometry. Mol Oncol 2025; 19:37-55. [PMID: 39234921 PMCID: PMC11705735 DOI: 10.1002/1878-0261.13723] [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: 04/06/2023] [Revised: 02/02/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024] Open
Abstract
In this paper, we have modified the workflow of the traditional light transmission aggregometry (LTA) protocol to characterize tumor cell clusters in vitro in a quantifiable and multifaceted manner. Circulating tumor cell (CTC) clusters have high metastatic potential compared to single tumor cells traveling in the bloodstream. Thus, engineering new therapeutic strategies that specifically target this CTC population is essential. To accomplish this, quantifiable methods to characterize their therapeutic effect on tumor cell clusters is a prerequisite. The method presented here enables the user to precisely quantify the dissociation of cancer cell clusters in the presence of clinically relevant fibrinolytic agents, such as alteplase and tenecteplase. The efficacy of the fibrinolytic agents can be quantified using this in vitro assay, prior to conducting preclinical studies. Here, we have obtained the fibrinolytic activity data in terms of lag time to the initiation of tumor cell dissociation, time to 25% dissociation, and trend of dissociation over time. To validate the assay, cell counts and phase-contrast microscopy images were recorded over time. Further, we explored an LTA-assisted preparation of platelet-tumor-cell clusters of calibrated size for potential downstream testing/applications. To assess whether the assay is applicable to characterize the dissociation of cancer cell clusters in the presence of platelets, we added low (50 000 platelets·μL-1), normal (200 000 platelets·μL-1) and high (450 000 platelets·μL-1) concentrations of platelets to the tumor cell clusters. In addition to dissociation parameters, microcopy images were recorded over time to validate the assay and enabled the enumeration of clusters and single cells. The correlative light electron microscopy (CLEM) technique was utilized to visualize the morphology and composition of platelet-tumor cell clusters.
Collapse
Affiliation(s)
- Regina Komal Kottana
- Department of Biomedical Engineering, School of Engineering and Applied ScienceThe George Washington UniversityWashingtonDCUSA
| | - Brian Schnoor
- Department of Biomedical Engineering, School of Engineering and Applied ScienceThe George Washington UniversityWashingtonDCUSA
| | - Anne‐Laure Papa
- Department of Biomedical Engineering, School of Engineering and Applied ScienceThe George Washington UniversityWashingtonDCUSA
| |
Collapse
|
10
|
Ünlü B, Heestermans M, Laghmani EH, Buijs JT, van den Akker RFP, van Vlijmen BJM, Versteeg HH. The effects of an aggressive breast tumor on thrombosis after antithrombin downregulation in a hypercoagulable mouse model. Thromb Res 2024; 244:109200. [PMID: 39476730 DOI: 10.1016/j.thromres.2024.109200] [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: 02/04/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024]
Abstract
BACKGROUND Despite improvements in therapy, breast cancer still contributes to high mortality rates. Survival of these patients becomes progressively worse upon diagnosis with cancer-associated thrombosis (CAT). Unfortunately, the mechanism causing CAT has remained unclear. OBJECTIVE Set up an acute and non-invasive hypercoagulable mouse model with an aggressive breast cancer and study the mechanism of cancer-associated thrombosis. METHODS Mice were grafted with the aggressive breast cancer cell line MDA-MB-231 or sham-treated. Subsequently, an acute imbalance in coagulation was introduced by injecting a synthetic small interfering (si) RNA targeting hepatic Serpinc1 to knockdown antithrombin - a condition known to predispose to cause a hypercoagulant state in vivo. RESULTS Silencing Serpinc1 with siRNA decreased plasma antithrombin levels. siRNA treatment had no short-term effects on tumor characteristics, but increased distant metastasis within the timeframe of this study. The systemic pro-inflammatory status, with elevated platelet counts and fibrinogen levels in tumor-bearing mice, was also not affected by antithrombin silencing. While elevated fibrin deposition in the liver upon Serpinc1 targeting was not significantly affected by the presence of breast cancer, knockdown of antithrombin did significantly increase intratumoral fibrin deposition and inflammation. Surprisingly, in the presence of an aggressive tumor, a protective outcome with less clinical features coinciding with venous thrombosis were observed in mice with antithrombin knockdown. CONCLUSION We conclude that the presence of a breast tumor protects hypercoagulant mice from severe consumption of coagulation factors after lowering hepatic antithrombin levels, possibly due to elevated platelet counts. However, the consequences on cancer-associated thrombosis remained inconclusive.
Collapse
Affiliation(s)
- Betül Ünlü
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Marco Heestermans
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - El Houari Laghmani
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob F P van den Akker
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Bart J M van Vlijmen
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis and Hemostasis, Department of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
11
|
Jomrich G, Yan W, Kollmann D, Kristo I, Winkler D, Puhr H, Lhan-Mutlu A, Hollenstein M, Asari R, Schoppmann SF. Elevated fibrinogen-albumin ratio is an adverse prognostic factor for patients with primarily resected gastroesophageal adenocarcinoma. J Cancer Res Clin Oncol 2024; 150:459. [PMID: 39400724 PMCID: PMC11473574 DOI: 10.1007/s00432-024-05976-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/25/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE Serum fibrinogen and albumin play important roles in systemic inflammation and are implicated in tumor progression. The fibrinogen-to-albumin ratio (FAR) has shown a prognostic impact in several malignancies. This study aims to assess the prognostic value of the pretherapeutic FAR in patients with adenocarcinoma of the gastroesophageal junction (AEG) who underwent upfront resection. METHODS Consecutive patients who underwent surgical resection at the Department of Surgery at the Medical University of Vienna between 1992 and 2014 were included into this study. Optimal cut-off values were determined with the receiver-operating characteristic (ROC) curve, uni- and multivariate analyzes were calculated by the Cox proportional hazard regression model for overall survival (OS). RESULTS Among 135 included patients, the majority were male (79.26%), with a mean age of 66.53 years. Elevated FAR correlated significantly (p = 0.002) with shorter OS in univariate analysis, also confirmed as independent prognostic factor (p = 0.005) in multivariable analysis. The ROC curve of FAR (AUC = 0.744) outperformed fibrinogen (AUC = 0.738) and albumin (AUC = 0.378) in predicting OS for AEG patients. CONCLUSION The FAR serves as an independent prognostic factor for OS in patients undergoing primarily resection for AEG. Given its routine availability and ease of calculation, FAR could help in diagnosis and treatment selection for AEG patients. Further validation studies are warranted to confirm these findings conclusively.
Collapse
Affiliation(s)
- Gerd Jomrich
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Winny Yan
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Dagmar Kollmann
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Ivan Kristo
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Daniel Winkler
- Vienna University of Economics and Business, Vienna, Austria
| | - Hannah Puhr
- Department of Medicine 1, Division of Oncology, Gastroesophageal Tumor Unit, Medical University of Vienna, Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Aysegül Lhan-Mutlu
- Department of Medicine 1, Division of Oncology, Gastroesophageal Tumor Unit, Medical University of Vienna, Comprehensive Cancer Center (CCC), Vienna, Austria
| | - Marlene Hollenstein
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Reza Asari
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria
| | - Sebastian F Schoppmann
- Department of General Surgery, Gastroesophageal Tumor Unit, Comprehensive Cancer Center (CCC), Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
12
|
Dobson DA, Fish RJ, de Vries PS, Morrison AC, Neerman-Arbez M, Wolberg AS. Regulation of fibrinogen synthesis. Thromb Res 2024; 242:109134. [PMID: 39216273 PMCID: PMC11381137 DOI: 10.1016/j.thromres.2024.109134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The plasma protein fibrinogen is encoded by 3 structural genes (FGA, FGB, and FGG) that are transcribed to mRNA, spliced, and translated to 3 polypeptide chains (Aα, Bβ, and γ, respectively). These chains are targeted for secretion, decorated with post-translational modifications, and assembled into a hexameric "dimer of trimers" (AαBβγ)2. Fully assembled fibrinogen is secreted into the blood as a 340 kDa glycoprotein. Fibrinogen is one of the most prevalent coagulation proteins in blood, and its expression is induced by inflammatory cytokines, wherein circulating fibrinogen levels may increase up to 3-fold during acute inflammatory events. Abnormal levels of circulating fibrinogen are associated with bleeding and thrombotic disorders, as well as several inflammatory diseases. Notably, therapeutic strategies to modulate fibrinogen levels have shown promise in experimental models of disease. Herein, we review pathways mediating fibrinogen synthesis, from gene expression to secretion. Knowledge of these mechanisms may lead to the identification of biomarkers and new therapeutic targets to modulate fibrinogen in health and disease.
Collapse
Affiliation(s)
- Dre'Von A Dobson
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, NC, USA
| | - Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Paul S de Vries
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, The University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
13
|
Ma C, Yang B, Mao Q. Prognostic value of fibrinogen change value in adrenocortical carcinoma patients. Discov Oncol 2024; 15:320. [PMID: 39080085 PMCID: PMC11289202 DOI: 10.1007/s12672-024-01197-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
PURPOSE The aim was to explore the preoperative and postoperative fibrinogen changes value (FCV) as a prognosis biomarker for in patients with adrenocortical carcinoma (ACC). METHODS We identified 42 patients with ACC and 190 patients with adrenal adenoma (AA) who underwent surgery at our institution between 2015 and 2023. Preoperative fibrinogen, postoperative fibrinogen and follow-up information of the patients were recorded and analysed. The relationship between FCV and overall survival (OS)/ relapse-free survival (RFS) was evaluated. RESULTS The mean level of preoperative and postoperative fibrinogen for ACC were 4.00 ± 1.64 g/L and 2.75 ± 0.59 g/L, respectively (p < 0.001). The mean level of preoperative and postoperative fibrinogen for AA were 2.79 ± 0.59 g/L and 2.71 ± 0.58 g/L, respectively (p = 0.144). In ACC, the lower FCV (≤ 1.25 g/L) showed a significantly poorer RFS than the higher (> 1.25 g/L) (p = 0.007); however, the lower FCV (≤ 1.25 g/L) showed no poorer OS than the higher (> 1.25 g/L) (p = 0.243). On multivariate survival analyses, FCV remained a predictor of RFS (HR 3.138). CONCLUSION According to the data in this study, it can be said that FCV is correlated with prognosis of ACC. The FCV might be a new biomarker for predicting the RFS of ACC.
Collapse
Affiliation(s)
- Chengquan Ma
- Department of Urology, Tianjin Medical University General Hospital, No 154. Anshan Road, Tianjin, 300052, China.
| | - Bin Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Quanzong Mao
- Department of Urology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| |
Collapse
|
14
|
Han X, Liu Z, Cui M, Lin J, Li Y, Qin H, Sheng J, Zhang X. FGA influences invasion and metastasis of hepatocellular carcinoma through the PI3K/AKT pathway. Aging (Albany NY) 2024; 16:12806-12819. [PMID: 39227068 PMCID: PMC11501378 DOI: 10.18632/aging.206011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 06/03/2024] [Indexed: 09/05/2024]
Abstract
Fibrinogen is an important plasma protein composed of three polypeptide chains, fibrinogen alpha (FGA), beta, and gamma. Apart from being an inflammation regulator, fibrinogen also plays a role in tumor progression. Liver cancer usually has a poor prognosis, with chronic hepatitis being the main cause of liver cirrhosis and hepatocellular carcinoma (HCC). FGA serves as a serological marker for chronic hepatitis, but its relationship with liver cancer remains unclear. Through bioinformatics analysis and agarose gel electrophoresis, we found that FGA was downregulated in HCC and correlated with tumor stage and grade. By constructing both FGA gene knockout and overexpression cell models, we demonstrated that overexpressing FGA inhibited migration and invasion of liver cancer cells through Transwell migration/invasion and wound healing assays. Western blotting experiments showed that FGA overexpression increased the expression of the epithelial-mesenchymal transition marker protein E-cadherin while decreasing N-cadherin and slug protein expression. In addition, FGA knockout activated the PI3K/AKT pathway. In a mouse model of metastatic tumors, overexpression of FGA restricted the spread of tumor cells. In conclusion, FGA exhibits an inhibitory effect on tumor metastasis, providing new insights for the treatment of advanced HCC metastatic tumors.
Collapse
Affiliation(s)
- Xi Han
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Zefeng Liu
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Mengying Cui
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Jie Lin
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Yongzhi Li
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Hanjiao Qin
- Department of Radiotherapy, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, Jilin 130041, China
| |
Collapse
|
15
|
Wang X, Wei W, Hua N, Li C, Yu L. Diagnostic value of serum fibrin degradation complex DR-70 combined with conventional tumor biomarkers in colorectal cancer. J Int Med Res 2024; 52:3000605241266236. [PMID: 39079131 PMCID: PMC11295240 DOI: 10.1177/03000605241266236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/13/2024] [Indexed: 08/04/2024] Open
Abstract
OBJECTIVE Most patients with colorectal cancer (CRC) show no early symptoms, and tumor markers have low sensitivity and specificity. We therefore investigated the ability of serum fibrin degradation complex DR-70 plus traditional tumor markers to diagnose CRC. METHODS We retrospectively screened patients with CRC or non-malignant colorectal diseases, as well as healthy individuals, for inclusion in this study. The individuals' clinical characteristics were recorded, and serum samples were collected. Expression levels of DR-70 and conventional tumor markers were measured by enzyme-linked immunosorbent assay and electrochemiluminescence. RESULTS DR-70 levels differed significantly among patients with CRC, patients with benign colorectal diseases, and healthy individuals. Receiver operating characteristic curve analysis identified DR-70 as a conventional tumor marker with the highest sensitivity and the second-highest specificity after carcinoembryonic antigen. CONCLUSIONS This study identified DR-70 as a reliable marker for the detection, differentiation, and progression of CRC, with good sensitivity and specificity. DR-70 measurement could greatly improve the efficacy of CRC diagnosis when used together with other tumor markers.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Hematology, Dongyang People’s Hospital, Zhejiang, China
| | - Wei Wei
- Postgraduate Training Base of Jinzhou Medical University (Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Nanni Hua
- Department of Nephrology, Hangzhou Traditional Chinese Medicine Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Chunyan Li
- Department of Oncology, Xuancheng City Central Hospital, Anhui, China
| | - Lili Yu
- Cancer Center, Department of Pathology, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Zhejiang, China
| |
Collapse
|
16
|
Zhu L, Liu S, Wang D, Yu M, Cai H. Relationship Between Coagulation and Prognosis of Gastric Cancer: A Systematic Review and Meta-Analysis. CURRENT THERAPEUTIC RESEARCH 2024; 101:100741. [PMID: 39628767 PMCID: PMC11612816 DOI: 10.1016/j.curtheres.2024.100741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 02/21/2024] [Indexed: 12/06/2024]
Abstract
Background The hypercoagulable state of cancer patients is associated with their high mortality rate. Coagulation indicators may have an important role in the prognosis of gastric cancer patients and deserve to be explored in various aspects. Objective We conducted a meta-analysis to explore the correlation between coagulation and prognosis of gastric cancer. Methods A comprehensive systematic search was conducted in PubMed, Embase, Web of Science databases, and the Cochrane Library up to February 16, 2024. Literature screening and data extraction were performed by two independent reviewers. The processed data we pooled using either a random-effects model or a fixed-effects model and finally described overall survival with a risk ratio (hazard ratio [HR]) and predicted the likelihood of different clinicopathological events with a dominance ratio (OR). Results A total of 64 studies were screened for inclusion in the data analysis. Performing a meta-analysis of three indicators we derived that the risk of d-dimer (D-D), fibrinogen (FIB), and platelets (PLTs) were: HR = 1.85 (95% confidence interval [CI]: 1.59-2.15, N = 15), HR = 1.77 (95% CI: 1.57-1.99, N = 28), HR = 1.16 (95% CI: 1.12-1.21, N = 29). In addition to this, all three were associated with advanced clinicopathological stage (D-D: OR = 2.25, FIB: OR = 2.07, PLT: OR = 1.84), T stage (D-D: OR = 2.30, FIB: OR = 2.38, PLT: OR = 2.22) and lymph node metastasis (D-D: OR = 1.79, FIB: OR = 1.70, PLT: OR = 1.51). Conclusion Overall, the findings suggest that the three indicators, D-D, FIB, and PLT count, have significant predictive value for the prognosis of gastric cancer. They were associated with an advanced clinicopathological stage and a high risk of lymph node metastasis.
Collapse
Affiliation(s)
- Lihui Zhu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor Gansu Provincial Hospital, Lanzhou, Gansu, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Shuo Liu
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor Gansu Provincial Hospital, Lanzhou, Gansu, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Da Wang
- Medical College of Jiangsu University, Zhenjiang, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor Gansu Provincial Hospital, Lanzhou, Gansu, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Miao Yu
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor Gansu Provincial Hospital, Lanzhou, Gansu, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Hui Cai
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
- Medical College of Jiangsu University, Zhenjiang, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor Gansu Provincial Hospital, Lanzhou, Gansu, China
- Gansu key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
17
|
Cornish N, Haycock P, Brenner H, Figueiredo JC, Galesloot TE, Grant RC, Johansson M, Mariosa D, McKay J, Pai R, Pellatt AJ, Samadder NJ, Shi J, Thibord F, Trégouët DA, Voegele C, Thirlwell C, Mumford A, Langdon R, InterLymph Consortium, INVENT-MVP Consortium. Causal relationships between risk of venous thromboembolism and 18 cancers: a bidirectional Mendelian randomization analysis. Int J Epidemiol 2024; 53:dyad170. [PMID: 38124529 PMCID: PMC10859161 DOI: 10.1093/ije/dyad170] [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: 05/16/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND People with cancer experience high rates of venous thromboembolism (VTE). Risk of subsequent cancer is also increased in people experiencing their first VTE. The causal mechanisms underlying this association are not completely understood, and it is unknown whether VTE is itself a risk factor for cancer. METHODS We used data from large genome-wide association study meta-analyses to perform bidirectional Mendelian randomization analyses to estimate causal associations between genetic liability to VTE and risk of 18 different cancers. RESULTS We found no conclusive evidence that genetic liability to VTE was causally associated with an increased incidence of cancer, or vice versa. We observed an association between liability to VTE and pancreatic cancer risk [odds ratio for pancreatic cancer: 1.23 (95% confidence interval: 1.08-1.40) per log-odds increase in VTE risk, P = 0.002]. However, sensitivity analyses revealed this association was predominantly driven by a variant proxying non-O blood group, with inadequate evidence to suggest a causal relationship. CONCLUSIONS These findings do not support the hypothesis that genetic liability to VTE is a cause of cancer. Existing observational epidemiological associations between VTE and cancer are therefore more likely to be driven by pathophysiological changes which occur in the setting of active cancer and anti-cancer treatments. Further work is required to explore and synthesize evidence for these mechanisms.
Collapse
Affiliation(s)
- Naomi Cornish
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Philip Haycock
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jane C Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Tessel E Galesloot
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert C Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Daniela Mariosa
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Rish Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Andrew J Pellatt
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Florian Thibord
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, USA
| | | | - Catherine Voegele
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Chrissie Thirlwell
- University of Exeter Medical School, University of Exeter, Exeter, UK
- Bristol Medical School, University of Bristol, Bristol, UK
| | - Andrew Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Ryan Langdon
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | | |
Collapse
|
18
|
Adnani L, Rak J. Intercellular Molecular Transfer Mediated by Extracellular Vesicles in Cancer. Results Probl Cell Differ 2024; 73:327-352. [PMID: 39242385 DOI: 10.1007/978-3-031-62036-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Among multiple pathways of intercellular communication operative in multicellular organisms, the trafficking of extracellular vesicles (EVs) and particles (EP) represents a unique mode of cellular information exchange with emerging roles in health and disease, including cancer. A distinctive feature of EV/EP-mediated cell-cell communication is that it involves simultaneous short- or long-range transfer of numerous molecular constituents (cargo) from donor to recipient cells. EV/EP uptake by donor cells elicits signalling or metabolic responses, or else leads to EV-re-emission or degradation. EVs are heterogeneous membranous structures released from cells via increasingly defined mechanisms involving either formation of multivesicular endosomes (exosomes) or budding from the plasma membrane (ectosomes). EPs (exomeres, supermeres) are membraneless complex particles, smaller than EVs and of less defined biogenesis and function. EVs/EPs carry complex assemblies of proteins, lipids and nucleic acids (RNA, DNA), which they shuttle into intercellular milieu, body fluids and recipient cells, via surface contact, fusion and different forms of internalization (endocytosis, micropinocytosis). While the physiological functions of EVs/EPs communication pathways continue to be investigated, their roles in cancer are increasingly well-defined. For example, EVs are involved in the transmission of cancer-specific molecular cargo, including mutant, oncogenic, transforming, or regulatory macromolecules to indolent, or normal cells, sometimes triggering their quasi-transformation-like states, or phenotypic alterations. Conversely, a reciprocal and avid uptake of stromal EVs by cancer cells may be responsible for modulating their oncogenic repertoire, as exemplified by the angiocrine effects of endothelial EVs influencing cancer cell stemness. EV exchanges during cancer progression have also been implicated in the formation of tumour stroma, angiogenesis and non-angiogenic neovascularization processes, immunosuppression, colonization of metastatic organ sites (premetastatic niche), paraneoplastic and systemic pathologies (thrombosis, diabetes, hepatotoxicity). Thus, an EV/EP-mediated horizontal transfer of cellular content emerges as a new dimension in cancer pathogenesis with functional, diagnostic, and therapeutic implications.
Collapse
Affiliation(s)
- Lata Adnani
- The Research Institute of the McGill University Health Centre, McGill University, QC, Canada
| | - Janusz Rak
- The Research Institute of the McGill University Health Centre, McGill University, QC, Canada.
| |
Collapse
|
19
|
Chowdhury NN, Yang Y, Dutta A, Luo M, Wei Z, Abrahams SR, Revenko AS, Shah F, Miles LA, Parmer RJ, de Laat B, Wolberg AS, Luyendyk JP, Fishel ML, Flick MJ. Plasminogen deficiency suppresses pancreatic ductal adenocarcinoma disease progression. Mol Oncol 2024; 18:113-135. [PMID: 37971174 PMCID: PMC10766200 DOI: 10.1002/1878-0261.13552] [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: 12/13/2022] [Revised: 10/06/2023] [Accepted: 11/14/2023] [Indexed: 11/19/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly fatal metastatic disease associated with robust activation of the coagulation and fibrinolytic systems. However, the potential contribution of the primary fibrinolytic protease plasminogen to PDAC disease progression has remained largely undefined. Mice bearing C57Bl/6-derived KPC (KRasG12D , TRP53R172H ) tumors displayed evidence of plasmin activity in the form of high plasmin-antiplasmin complexes and high plasmin generation potential relative to mice without tumors. Notably, plasminogen-deficient mice (Plg- ) had significantly diminished KPC tumor growth in subcutaneous and orthotopic implantation models. Moreover, the metastatic potential of KPC cells was significantly diminished in Plg- mice, which was linked to reduced early adhesion and/or survival of KPC tumor cells. The reduction in primary orthotopic KPC tumor growth in Plg- mice was associated with increased apoptosis, reduced accumulation of pro-tumor immune cells, and increased local proinflammatory cytokine production. Elimination of fibrin(ogen), the primary proteolytic target of plasmin, did not alter KPC primary tumor growth and resulted in only a modest reduction in metastatic potential. In contrast, deficiencies in the plasminogen receptors Plg-RKT or S100A10 in tumor cells significantly reduced tumor growth. Plg-RKT reduction in tumor cells, but not reduced S100A10, suppressed metastatic potential in a manner that mimicked plasminogen deficiency. Finally, tumor growth was also reduced in NSG mice subcutaneously or orthotopically implanted with patient-derived PDAC tumor cells in which circulating plasminogen was pharmacologically reduced. Collectively, these studies suggest that plasminogen promotes PDAC tumor growth and metastatic potential, in part through engaging plasminogen receptors on tumor cells.
Collapse
Affiliation(s)
- Nayela N. Chowdhury
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
| | - Yi Yang
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Ananya Dutta
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Michelle Luo
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - Zimu Wei
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Sara R. Abrahams
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | | | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
| | - Lindsey A. Miles
- Department of Molecular MedicineScripps Research InstituteLa JollaCAUSA
| | - Robert J. Parmer
- Department of Medicine, Veterans Administration San Diego Healthcare SystemUniversity of California, San DiegoCAUSA
| | - Bas de Laat
- Synapse Research InstituteMaastrichtThe Netherlands
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| | - James P. Luyendyk
- Department of Pathobiology & Diagnostic InvestigationMichigan State UniversityEast LansingMIUSA
- Institute for Integrative ToxicologyMichigan State UniversityEast LansingMIUSA
- Department of Pharmacology and ToxicologyMichigan State UniversityEast LansingMIUSA
| | - Melissa L. Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric ResearchIndianapolisINUSA
- Indiana University Simon Comprehensive Cancer CenterIndianapolisINUSA
- Department of Pharmacology and ToxicologyIndiana University School of MedicineIndianapolisINUSA
| | - Matthew J. Flick
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillNCUSA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina at Chapel HillNCUSA
- UNC Blood Research CenterUniversity of North Carolina at Chapel HillNCUSA
| |
Collapse
|
20
|
Baghaie L, Haxho F, Leroy F, Lewis B, Wawer A, Minhas S, Harless WW, Szewczuk MR. Contemporaneous Perioperative Inflammatory and Angiogenic Cytokine Profiles of Surgical Breast, Colorectal, and Prostate Cancer Patients: Clinical Implications. Cells 2023; 12:2767. [PMID: 38067195 PMCID: PMC10706122 DOI: 10.3390/cells12232767] [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: 11/08/2023] [Revised: 11/29/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Surgery-induced tumor growth acceleration and synchronous metastatic growth promotion have been observed for decades. Surgery-induced wound healing, orchestrated through growth factors, chemokines, and cytokines, can negatively impact patients harboring residual or metastatic disease. We provide detailed clinical evidence of this process in surgical breast, prostate, and colorectal cancer patients. Plasma samples were analyzed from 68 cancer patients who had not received treatment before surgery or adjuvant therapy until at least four weeks post-surgery. The levels of plasma cytokines, chemokines, and growth factors were simultaneously quantified and profiled using multiplexed immunoassays for eight time points sampled per patient. The immunologic processes are induced immediately after surgery in patients, characterized by a drastic short-term shift in the expression levels of pro-inflammatory and angiogenic molecules and cytokines. A rapid and significant spike in circulating plasma levels of hepatocyte growth factor (HGF), interleukin-6 (IL-6), placental growth factor (PLGF), and matrix metalloproteinase-9 (MMP-9) after surgery was noted. The rise in these molecules was concomitant with a significant drop in transforming growth factor-β1 (TGF-β1), platelet-derived growth factor (PDGF-AB/BB), insulin-like growth factor-1 (IGF-1), and monocyte chemoattractant protein-2 (MCP-2). If not earlier, each plasma analyte was normalized to baseline levels within 1-2 weeks after surgery, suggesting that surgical intervention alone was responsible for these effects. The effects of surgical tumor removal on disrupting the pro-inflammatory and angiogenic plasma profiles of cancer patients provide evidence for potentiating malignant progression. Our findings indicate a narrow therapeutic window of opportunity after surgery to prevent disease recurrence.
Collapse
Affiliation(s)
- Leili Baghaie
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
| | - Fiona Haxho
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
- Dermatology Residency Program, the Cumming School of Medicine, University of Calgary, Calgary, AB T2T 5C7, Canada
| | - Fleur Leroy
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
- Faculté de Médecine, Maïeutique et Sciences de la Santé, Université de Strasbourg, F-67000 Strasbourg, France
| | - Beth Lewis
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Alexander Wawer
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Shamano Minhas
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - William W. Harless
- ENCYT Technologies Inc., Membertou, NS B1S 0H1, Canada; (B.L.); (A.W.); (S.M.)
| | - Myron R. Szewczuk
- Department of Biomedical & Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (L.B.); (F.H.); (F.L.)
| |
Collapse
|
21
|
Matovinovic F, Novak R, Hrkac S, Salai G, Mocibob M, Pranjic M, Košec A, Bedekovic V, Grgurevic L. In search of new stratification strategies: tissue proteomic profiling of papillary thyroid microcarcinoma in patients with localized disease and lateral neck metastases. J Cancer Res Clin Oncol 2023; 149:17405-17417. [PMID: 37861757 DOI: 10.1007/s00432-023-05452-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/30/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION Papillary thyroid carcinomas (PTC) are the most common thyroid malignancies that are often diagnosed as microcarcinomas when the tumor is less than one centimetre in diameter. Currently, there are no valid stratification strategies that would reliably assess the risk of lateral neck metastases and optimize surgical treatment. MATERIALS AND METHODS Aiming to find potential tissue biomarkers of metastatic potential, we conducted a cross-sectional proteomic pilot study on formalin-fixed paraffin-embedded tissues of metastatic (N = 10) and non-metastatic (N = 10) papillary thyroid microcarcinoma patients. Samples were analysed individually using liquid chromatography/mass spectrometry, and the differentially expressed proteins (DEP) were functionally annotated. RESULTS We identified five overexpressed DEPs in the metastatic group (EPB41L2, CSE1L, GLIPR2, FGA and FGG) with a known association to tumour biology. Using bioinformatic-based tools, we found markedly different profiles of significantly enriched biological processes between the two groups. CONCLUSIONS The identified DEPs might have a role as potential tissue biomarkers for PTC metastases. However, further prospective research is needed to confirm our findings.
Collapse
Affiliation(s)
- Filip Matovinovic
- Department of Otorhinolaryngology and Head and Neck Surgery, Sestre Milosrdnice University Hospital Center, 10000, Zagreb, Croatia
| | - Rudjer Novak
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Stela Hrkac
- Department of Clinical Immunology, Allergology and Rheumatology, University Hospital Dubrava, 10000, Zagreb, Croatia
| | - Grgur Salai
- Department of Pulmonology, University Hospital Dubrava, 10000, Zagreb, Croatia
| | - Marko Mocibob
- Department of Chemistry, Faculty of Science, University of Zagreb, 10000, Zagreb, Croatia
| | - Marija Pranjic
- Department of Chemistry, Faculty of Science, University of Zagreb, 10000, Zagreb, Croatia
| | - Andro Košec
- Department of Otorhinolaryngology and Head and Neck Surgery, Sestre Milosrdnice University Hospital Center, 10000, Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000, Zagreb, Croatia
| | - Vladimir Bedekovic
- Department of Otorhinolaryngology and Head and Neck Surgery, Sestre Milosrdnice University Hospital Center, 10000, Zagreb, Croatia
| | - Lovorka Grgurevic
- Center for Translational and Clinical Research, Department of Proteomics, School of Medicine, University of Zagreb, 10000, Zagreb, Croatia.
- Department of Anatomy, "Drago Perovic", School of Medicine, University of Zagreb, 10000, Zagreb, Croatia.
| |
Collapse
|
22
|
Heilala M, Lehtonen A, Arasalo O, Peura A, Pokki J, Ikkala O, Nonappa, Klefström J, Munne PM. Fibrin Stiffness Regulates Phenotypic Plasticity of Metastatic Breast Cancer Cells. Adv Healthc Mater 2023; 12:e2301137. [PMID: 37671812 PMCID: PMC11469292 DOI: 10.1002/adhm.202301137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/18/2023] [Indexed: 09/07/2023]
Abstract
The extracellular matrix (ECM)-regulated phenotypic plasticity is crucial for metastatic progression of triple negative breast cancer (TNBC). While ECM faithful cell-based models are available for in situ and invasive tumors, such as cell aggregate cultures in reconstituted basement membrane and in collagenous gels, there are no ECM faithful models for metastatic circulating tumor cells (CTCs). Such models are essential to represent the stage of metastasis where clinical relevance and therapeutic opportunities are significant. Here, CTC-like DU4475 TNBC cells are cultured in mechanically tunable 3D fibrin hydrogels. This is motivated, as in circulation fibrin aids CTC survival by forming a protective coating reducing shear stress and immune cell-mediated cytotoxicity and promotes several stages of late metastatic processes at the interface between circulation and tissue. This work shows that fibrin hydrogels support DU4475 cell growth, resulting in spheroid formation. Furthermore, increasing fibrin stiffness from 57 to 175 Pa leads to highly motile, actin and tubulin containing cellular protrusions, which are associated with specific cell morphology and gene expression patterns that markedly differ from basement membrane or suspension cultures. Thus, mechanically tunable fibrin gels reveal specific matrix-based regulation of TNBC cell phenotype and offer scaffolds for CTC-like cells with better mechano-biological properties than liquid.
Collapse
Affiliation(s)
- Maria Heilala
- Department of Applied PhysicsAalto UniversityP.O. Box 15100AaltoEspooFI‐00076Finland
| | - Arttu Lehtonen
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Ossi Arasalo
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Aino Peura
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| | - Juho Pokki
- Department of Electrical Engineering and AutomationAalto UniversityP.O. Box 12200AaltoEspooFI‐00076Finland
| | - Olli Ikkala
- Department of Applied PhysicsAalto UniversityP.O. Box 15100AaltoEspooFI‐00076Finland
| | - Nonappa
- Faculty of Engineering and Natural SciencesTampere UniversityP.O. Box 541TampereFI‐33720Finland
| | - Juha Klefström
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| | - Pauliina M. Munne
- Finnish Cancer Institute and FICAN SouthHelsinki University Hospital & Cancer Cell Circuitry LaboratoryTranslational Cancer MedicineMedical FacultyUniversity of HelsinkiP.O. Box 63 (Haartmaninkatu 8)Helsinki00014Finland
| |
Collapse
|
23
|
Tian H, Liu Z, Zhang Z, Zhang L, Zong Z, Liu J, Ying H, Li H. Clinical Significance of Fibrinogen and Platelet to Pre-Albumin Ratio in Predicting the Prognosis of Advanced Gastric Cancer. J Inflamm Res 2023; 16:4373-4388. [PMID: 37808954 PMCID: PMC10557981 DOI: 10.2147/jir.s412033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/13/2023] [Indexed: 10/10/2023] Open
Abstract
OBJECTIVE The aim of this study was to investigate the clinical significance of Fibrinogen and Platelet to Pre-albumin Ratio(FPAR) in predicting the prognosis of patients with advanced gastric cancer(AGC) and to construct a predictive model. METHODS We collected clinical data from 489 postoperative patients with AGC. FPAR was divided into high and low groups according to the receiver operating characteristic (ROC) curve. The value of FPAR in predicting the prognosis of progressive gastric cancer was analysed using univariate and multivariable Cox regression analysis and its relationship with clinicopathological features. Finally, the Overall Survival(OS) and recurrence-free survival(RFS) prediction models were constructed and validated using FPAR. RESULTS Univariate and multifactorial cox regression analysis showed that grade (P<0.001), TNM-stage (P<0.001), chemotherapy (P<0.001), and FPAR (OR=3.054,95% CI:2.088-4.467, P<0.001) were independent risk factors for OS; grade (P=0.021), N-stage (P=0.024), TNM-stage (P=0.033), and FPAR (OR=2.215,95% CI:1.634-3.003, P<0.001) were independent risk factors for RFS. Subgroup analysis showed that the FPAR-low group had higher OS and RFS than the FPAR-high group, regardless of the patient's TNM stage (p<0.05). However, OS was instead higher in the the stage III-FPAR-low group than in the the stage II-FPAR-high group (p<0.05), while RFS was not significantly different. Predictive models incorporating FPAR had better predictive performance than those without FPAR, showing wide range of net benefit and AUC. After correction, the 2-year AUC, 3-year AUC and C-index of the OS model were 0.737, 0.756, and 0.746; the 2-year AUC, 3-year AUC, and C-index of the RFS model were 0.738, 0.758, and 0.711. CONCLUSION FPAR levels were associated with prognosis in patients with AGC and could independently predict RFS and OS.
Collapse
Affiliation(s)
- Huakai Tian
- Department of General Surgery, First Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zitao Liu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zuo Zhang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Lipeng Zhang
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Jiang Liu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Houqun Ying
- Department of Nuclear Medicine, Jiangxi Province Key Laboratory of Laboratory Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Hui Li
- Department of Rheumatology and Immunology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| |
Collapse
|
24
|
Li F, Ren Y, Fan J, Zhou J. The predictive value of the preoperative albumin-to-fibrinogen ratio for postoperative hospital length of stay in liver cancer patients. Cancer Med 2023; 12:20321-20331. [PMID: 37815011 PMCID: PMC10652297 DOI: 10.1002/cam4.6606] [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: 06/02/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a significant global health burden, with postoperative hospital length of stay (LOS) impacting patient outcomes and healthcare costs. Existing nutritional, inflammatory, and coagulation indices can predict LOS, with particular interest in albumin, fibrinogen, and D-dimer. This study investigates the predictive value of preoperative albumin-to-fibrinogen ratio (AFR) and albumin-to-D-dimer ratio (ADR) for postoperative LOS in HCC patients. METHODS This retrospective study involved 462 adult HCC patients who underwent partial hepatic lesion excision between February 2016 and August 2022. We analyzed demographic and clinical data, including preoperative blood samples, surgical approach, and LOS. The primary outcome measure was LOS, calculated from the date of surgery to the date of hospital discharge. Preoperative AFR and ADR were calculated. The ROC curves determined optimal cutoff points. The Cox proportional hazards model, Kaplan-Meier method, and the log-rank test were used for statistical analysis. RESULTS The study established an optimal AFR cutoff value of 15.474, with a higher AUC value than ADR, indicating superior predictive potential for postoperative LOS. Participants with high-AFR (AFR > 15.474) had a shorter median LOS (13 vs. 15 days, p < 0.001) compared to those with low-AFR (AFR ≤15.474). Multivariate analysis revealed high-AFR (HR: 1.99; p < 0.001) as a positive influence on LOS reduction, whereas Child-Pugh rated as B (HR: 0.49; p < 0.001), laparotomy (HR: 0.37; p < 0.001) and total bilirubin >20.5 μmol/L (HR: 0.58; p < 0.001) negatively impacted LOS reduction. Subgroup analysis confirmed AFR's predictive ability for patients experiencing reduced or prolonged LOS due to Child-Pugh score, surgical methods, and total bilirubin concentrations. Even within normal albumin and fibrinogen levels, patients with high-AFR exhibited a shorter LOS (all p < 0.001). CONCLUSIONS Our findings underscore the value of the AFR as a reliable predictor of LOS in HCC patients. An AFR greater than 15.474 consistently correlated with a shorter LOS, suggesting its potential clinical utility in guiding perioperative management and resource allocation in HCC patients.
Collapse
Affiliation(s)
- Fang Li
- Department of Hepatobiliary SurgeryLiaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yuetong Ren
- Department of Hepatobiliary SurgeryLiaoning Cancer Hospital & Institute, Cancer Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiacheng Fan
- Department of Medical Laboratory Technology, Medical SchoolShandong Xiandai UniversityJinanShandongChina
| | - Jin Zhou
- Medical Oncology Department of Gastrointestinal CancerLiaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of TechnologyLiaoningShenyangChina
| |
Collapse
|
25
|
Lei X, Zhang T, Deng Z, Jiang T, Hu Y, Yang N. Coagulation markers as independent predictors of prostate cancer aggressiveness: a retrospective cohort study. Sci Rep 2023; 13:16073. [PMID: 37752191 PMCID: PMC10522718 DOI: 10.1038/s41598-023-43427-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: 04/11/2023] [Accepted: 09/23/2023] [Indexed: 09/28/2023] Open
Abstract
Coagulation system activation is commonly observed in tumor patients, including prostate cancer (PCa), with coagulation markers proposed as potential prognostic indicators for cancer severity. However, the correlation between these markers and clinicopathological features in PCa remains unclear. Thus, this study investigates the association between comprehensive coagulation markers and clinicopathological characteristics in PCa patients. A retrospective evaluation of 162 PCa patients diagnosed and categorized into low-intermediate-risk or high-risk groups based on clinical and pathological features was conducted. Coagulation markers, including fibrinogen (FIB), D-dimer (DD), activated partial thromboplastin time (APTT), prothrombin time (PT), prothrombin activity (PTA), thrombin time (TT), platelet count (PLT), and international normalized ratio (INR), were assessed. Univariate and multivariate logistic regression analyses were performed to determine associations with clinicopathological features. FIB and DD were confirmed as independent factors associated with high-risk PCa. Furthermore, FIB and DD levels showed significant positive correlations with clinical parameters, including PSA levels, ISUP grade, T stage, N stage, and M stage. Our findings suggest that FIB and DD hold promise as independent prognostic biomarkers for risk stratification in PCa. These coagulation markers may aid in assessing PCa severity and guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Xu Lei
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Tengfei Zhang
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Zhixuan Deng
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Tao Jiang
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Yang Hu
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China
| | - Ning Yang
- The Second Affiliated Hospital of University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
26
|
Gois GSS, Montalvão SAL, Anhaia TRA, Almeida MEA, Martinelli BM, Fernandes MCGL, Hubers SC, Ferreira MRM, Ribeiro DD, Teixeira JC, Carvalheira JBC, Lima CSP, Andreollo NA, Etchebehere M, Zambon L, Ferreira U, Tincani AJ, Martins AS, Coy CSR, Seabra JCT, Mussi RK, Tedeschi H, Anninchino-Bizzacchi JM, ADVENTH Cancer Group. Association of Fibrinolytic Potential and Risk of Mortality in Cancer Patients. Cancers (Basel) 2023; 15:4408. [PMID: 37686683 PMCID: PMC10487037 DOI: 10.3390/cancers15174408] [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: 06/21/2023] [Revised: 08/02/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Cancer is a leading cause of death, and the fibrinolytic system shows cooperative effects that facilitate the growth of tumors and the appearance of metastases. This prospective study aimed to evaluate the fibrinolytic potential in cancer patients and its association with mortality outcomes using the fluorometric method of simultaneous thrombin and plasmin generation. The study included 323 cancer patients and 148 healthy individuals. During the 12-month follow-up, 68 patients died. Compared to the control group, cancer patients showed alterations in thrombin production consistent with a hypercoagulability profile, and an increase in plasmin generation. Mortality risk was associated with two parameters of thrombin in both univariate and multivariable analysis: maximum amplitude (Wald 11.78, p < 0.001) and area under the curve (Wald 8.0, p < 0.005), while such associations were not observed for plasmin. In conclusion, this was the first study able to demonstrate the simultaneous evaluation of thrombin and plasmin generation in newly diagnosed untreated cancer patients. Patients with cancer have been observed to exhibit a hypercoagulable profile. During the study, two parameters linked to thrombin generation, MA and AUC, were identified and found to have a potential association with mortality risk. However, no associations were found with parameters related to plasmin generation.
Collapse
Affiliation(s)
- Gabriele Silva Souza Gois
- School of Medical Science, FCM-UNICAMP, University of Campinas, Campinas 13083-888, SP, Brazil (J.M.A.-B.)
| | - Silmara Aparecida Lima Montalvão
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | | | - Millene Evelyn Alves Almeida
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | - Beatriz Moraes Martinelli
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | | | - Stephany Cares Hubers
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | - Monique R. M. Ferreira
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | | | - Júlio César Teixeira
- Department of Obstetrics and Gynecology, Division of Oncology, Women’s Hospital, CAISM-UNICAMP, University of Campinas, Campinas 13083-881, SP, Brazil
| | | | | | | | | | - Lair Zambon
- Clinical Hospital of Unicamp, Campinas 13083-888, SP, Brazil
| | | | | | | | | | | | | | - Helder Tedeschi
- Clinical Hospital of Unicamp, Campinas 13083-888, SP, Brazil
| | - Joyce Maria Anninchino-Bizzacchi
- School of Medical Science, FCM-UNICAMP, University of Campinas, Campinas 13083-888, SP, Brazil (J.M.A.-B.)
- Laboratory of Hemostasis, Hematology and Transfusion Medicine Center, Hemocentro—UNICAMP, Campinas 13083-878, SP, Brazil; (S.A.L.M.)
| | | |
Collapse
|
27
|
Li Q, Li L, Wang Y, Xu C, Zou J. The prognostic value of pretreatment albumin-to-fibrinogen ratio in small cell lung cancer patients receiving first-line platinum-based chemotherapy. Heliyon 2023; 9:e19225. [PMID: 37662747 PMCID: PMC10470268 DOI: 10.1016/j.heliyon.2023.e19225] [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: 05/18/2023] [Revised: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
This study examined the role of pretreatment albumin-to-fibrinogen ratio (AFR) in the prognosis of small-cell lung cancer (SCLC) patients receiving first-line platinum-based chemotherapy. A total of 131 SCLC patients were enrolled. The predictive value of the AFR for progression free survival (PFS) and overall survival (OS) were evaluated by receiver operating characteristic (ROC) curve analysis. The predictive factor of survival was assessed by univariate and multivariate Cox proportional regression analysis. The correlation between OS, PFS and AFR was determined by the log-rank test using the Kaplan-Meier method. AFR was an effective predictor of OS in SCLC patients with a cut-off value of 7.78. AFR was independent risk factors for OS and PFS. Kaplan Meier analysis showed that PFS and OS in patients with high AFR levels were significantly higher than those with low AFR levels. These results suggest that AFR could be an effective predictor of survival in patients with SCLC.
Collapse
Affiliation(s)
- Qi Li
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Li Li
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Yuchao Wang
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Chunhua Xu
- Department of Respiratory Medicine, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jue Zou
- Department of Pathology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| |
Collapse
|
28
|
Zhong S, Borlak J. Sex disparities in non-small cell lung cancer: mechanistic insights from a cRaf transgenic disease model. EBioMedicine 2023; 95:104763. [PMID: 37625265 PMCID: PMC10470261 DOI: 10.1016/j.ebiom.2023.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/10/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Women are at greater risk of developing non-small cell lung cancer (NSCLC), yet the underlying causes remain unclear. METHODS We performed whole genome scans in lung tumours of cRaf transgenic mice and identified miRNA, transcription factor and hormone receptor dependent gene regulations. We confirmed hormone receptors by immunohistochemistry and constructed regulatory gene networks by considering experimentally validated miRNA-gene and transcription factor-miRNA/gene targets. Bioinformatics, genomic foot-printing and gene enrichment analysis established sex-specific circuits of lung tumour growth. Translational research involved a large cohort of NSCLC patients. We evaluated commonalities in sex-specific NSCLC gene regulations between mice and humans and determined their prognostic value in Kaplan-Meier survival statistics and COX proportional hazard regression analysis. FINDINGS Overexpression of the cRaf kinase elicited an extraordinary 8-fold increase in tumour growth among females, and nearly 70% of the 112 differentially expressed genes (DEGs) were female specific. We identified oncogenes, oncomirs, tumour suppressors, cell cycle regulators and MAPK/EGFR signalling molecules, which prompted sex-based differences in NSCLC, and we deciphered a regulatory gene-network, which protected males from accelerated tumour growth. Strikingly, 41% of DEGs are targets of hormone receptors, and the majority (85%) are oestrogen receptor (ER) dependent. We confirmed the role of ER in a large cohort of NSCLC patients and validated 40% of DEGs induced by cRaf in clinical tumour samples. INTERPRETATION We report the molecular wiring that prompted sex disparities in tumour growth. This allowed us to propose the development of molecular targeted therapies by jointly blocking ER, CDK1 and arginase 2 in NSCLC. FUNDING We gratefully acknowledge the financial support of the Lower Saxony Ministry of Culture and Sciences and Volkswagen Foundation, Germany to JB (25A.5-7251-99-3/00) and of the Chinese Scholarship Council to SZ (202008080022). This publication is funded by the Deutsche Forschungsgemeinschaft (DFG) as part of the "Open Access Publikationskosten" program.
Collapse
Affiliation(s)
- Shen Zhong
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Carl-Neuberg-Str. 1, Hannover 30625, Germany.
| |
Collapse
|
29
|
Thaler J, Prager G, Pabinger I, Ay C. Plasma Clot Properties in Patients with Pancreatic Cancer. Cancers (Basel) 2023; 15:4030. [PMID: 37627058 PMCID: PMC10452192 DOI: 10.3390/cancers15164030] [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: 05/04/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic cancer is one of the most prothrombotic malignancies. Plasma clot properties may be altered in patients with pancreatic cancer, and circulating tissue factor (TF) may play an important role. We applied a modified plasma clot formation assay (only CaCl2 and phospholipids were added to initiate clotting) and a standard clotting assay (lipidated TF was also added) to investigate whether plasma clot properties are altered in pancreatic cancer patients (n = 40, 23 female) compared to sex-matched healthy controls. The modified assay was also performed in the presence of a TF blocking antibody. With this modified assay, we detected an increased plasma clot formation rate (Vmax) and an increased delta absorbance (ΔAbs, indicating fibrin fiber thickness) in patients compared to controls. These differences were not detected with the standard clotting assay. Following addition of a TF blocking antibody in in our modified assay, Vmax decreased significantly in patients only, ΔAbs significantly decreased in patients and in healthy controls, the lag phase did not change, and the time to peak fibrin generation increased in patients only. Taken together, these findings indicate the presence of a prothrombotic state in pancreatic cancer patients, which depends on TF and is detectable with our modified assay but not with a standard clotting assay.
Collapse
Affiliation(s)
- Johannes Thaler
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| | - Gerald Prager
- Clinical Division of Oncology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ingrid Pabinger
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Medical University of Vienna, 1090 Vienna, Austria; (I.P.); (C.A.)
| |
Collapse
|
30
|
Cao H, Shi H, Zhao M, Liu Z, Qian J. Prognostic value of the combined preoperative plasma fibrinogen and systemic inflammatory indexes in ESCC patients. Discov Oncol 2023; 14:143. [PMID: 37541963 PMCID: PMC10403484 DOI: 10.1007/s12672-023-00763-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023] Open
Abstract
The prognostic indexes based on the combination of preoperative fibrinogen and systemic inflammatory indexes may have greater predictive value in esophageal squamous cell carcinoma (ESCC). It was found that the predictive ability of F-NLR was more valuable than other systemic inflammatory indexes. The preoperative F-NLR score was closely related to the TNM stage, and could be used as an important independent prognostic index for patients with ESCC. Then the nomogram model constructed by F-NLR and TNM stage had higher prognostic ability than that of AJCC stage for ESCC patients. Preoperative F-NLR is a new independent prognostic index and a potential marker for treatment response monitoring in patients with ESCC.
Collapse
Affiliation(s)
- Honggang Cao
- Department of Oncology, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, 75 Juchang Street, Yancheng, 224005, China
| | - Hongtai Shi
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Nantong University, Yancheng Third People's Hospital, 75 Juchang Street, Yancheng, 224005, China
| | - Miaomiao Zhao
- Department of Ultrasound, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, 66 Renmin Road, Yancheng, 224005, China
| | - Zhenhua Liu
- Department of Radiotherapy, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, 66 Renmin Road, Yancheng, 224005, China.
| | - Jun Qian
- Department of Thoracic Surgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, 66 Renmin Road, Yancheng, 224005, China.
| |
Collapse
|
31
|
Angelidakis E, Chen S, Zhang S, Wan Z, Kamm RD, Shelton SE. Impact of Fibrinogen, Fibrin Thrombi, and Thrombin on Cancer Cell Extravasation Using In Vitro Microvascular Networks. Adv Healthc Mater 2023; 12:e2202984. [PMID: 37119127 PMCID: PMC10524192 DOI: 10.1002/adhm.202202984] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/25/2023] [Indexed: 04/30/2023]
Abstract
A bidirectional association exists between metastatic dissemination and the hypercoagulable state associated with many types of cancer. As such, clinical studies have provided evidence that markers associated with elevated levels of coagulation and fibrinolysis correlate with decreased patient survival. However, elucidating the mechanisms underpinning the effects of different components of the coagulation system on metastasis formation is challenging both in animal models and 2D models lacking the complex cellular interactions necessary to model both thrombosis and metastasis. Here, an in vitro, 3D, microvascular model for observing the formation of fibrin thrombi is described, which is in turn used to study how different aspects of the hypercoagulable state associated with cancer affect the endothelium. Using this platform, cancer cells expressing ICAM-1 are shown to form a fibrinogen-dependent bridge and transmigrate through the endothelium more effectively. Cancer cells are also demonstrated to interact with fibrin thrombi, using them to adhere, spread, and enhance their extravasation efficiency. Finally, thrombin is also shown to enhance cancer cell extravasation. This system presents a physiologically relevant model of fibrin clot formation in the human microvasculature, enabling in-depth investigation of the cellular interactions between cancer cells and the coagulation system affecting cancer cell extravasation.
Collapse
Affiliation(s)
- Emmanouil Angelidakis
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sophia Chen
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Shun Zhang
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Zhengpeng Wan
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Roger D. Kamm
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Sarah E. Shelton
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Medical OncologyDana Farber Cancer InstituteBostonMA02215USA
| |
Collapse
|
32
|
Rodriguez BL, Chen L, Li Y, Miao S, Peng DH, Fradette JJ, Diao L, Konen JM, Alvarez FRR, Solis LM, Yi X, Padhye A, Gibson LA, Ochieng JK, Zhou X, Wang J, Gibbons DL. Targeting immunosuppressive Ly6C+ classical monocytes reverses anti-PD-1/CTLA-4 immunotherapy resistance. Front Immunol 2023; 14:1161869. [PMID: 37449205 PMCID: PMC10336223 DOI: 10.3389/fimmu.2023.1161869] [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: 02/08/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Introduction Despite significant clinical advancement with the use of immune checkpoint blockade (ICB) in non-small cell lung cancer (NSCLC) there are still a major subset of patients that develop adaptive/acquired resistance. Understanding resistance mechanisms to ICB is critical to developing new therapeutic strategies and improving patient survival. The dynamic nature of the tumor microenvironment and the mutational load driving tumor immunogenicity limit the efficacy to ICB. Recent studies indicate that myeloid cells are drivers of ICB resistance. In this study we sought to understand which immune cells were contributing to resistance and if we could modify them in a way to improve response to ICB therapy. Results Our results show that combination anti-PD-1/CTLA-4 produces an initial antitumor effect with evidence of an activated immune response. Upon extended treatment with anti-PD-1/CTLA-4 acquired resistance developed with an increase of the immunosuppressive populations, including T-regulatory cells, neutrophils and monocytes. Addition of anti-Ly6C blocking antibody to anti-PD-1/CTLA-4 was capable of completely reversing treatment resistance and restoring CD8 T cell activity in multiple KP lung cancer models and in the autochthonous lung cancer KrasLSL-G12D/p53fl/fl model. We found that there were higher classical Ly6C+ monocytes in anti-PD-1/CTLA-4 combination resistant tumors. B7 blockade illustrated the importance of dendritic cells for treatment efficacy of anti-Ly6C/PD-1/CTLA-4. We further determined that classical Ly6C+ monocytes in anti-PD-1/CTLA-4 resistant tumors are trafficked into the tumor via IFN-γ and the CCL2-CCR2 axis. Mechanistically we found that classical monocytes from ICB resistant tumors were unable to differentiate into antigen presenting cells and instead differentiated into immunosuppressive M2 macrophages or myeloid-derived suppressor cells (MDSC). Classical Ly6C+ monocytes from ICB resistant tumors had a decrease in both Flt3 and PU.1 expression that prevented differentiation into dendritic cells/macrophages. Conclusions Therapeutically we found that addition of anti-Ly6C to the combination of anti-PD-1/CTLA-4 was capable of complete tumor eradication. Classical Ly6C+ monocytes differentiate into immunosuppressive cells, while blockade of classical monocytes drives dendritic cell differentiation/maturation to reinvigorate the anti-tumor T cell response. These findings support that immunotherapy resistance is associated with infiltrating monocytes and that controlling the differentiation process of monocytes can enhance the therapeutic potential of ICB.
Collapse
Affiliation(s)
- B. Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Limo Chen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Yanli Li
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shucheng Miao
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - David H. Peng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jared J. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lixia Diao
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jessica M. Konen
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Frank R. Rojas Alvarez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M. Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaohui Yi
- Bellicum Pharmaceuticals, Inc., Houston, TX, United States
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Aparna Padhye
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- United of Texas (UT) Health Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| | - Laura A. Gibson
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joshua K. Ochieng
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaofei Zhou
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Wang
- Department Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
33
|
Li S, Zhang H, Zhu M, Kuang Z, Li X, Xu F, Miao S, Zhang Z, Lou X, Li H, Xia F. Electrochemical Biosensors for Whole Blood Analysis: Recent Progress, Challenges, and Future Perspectives. Chem Rev 2023. [PMID: 37262362 DOI: 10.1021/acs.chemrev.1c00759] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Whole blood, as one of the most significant biological fluids, provides critical information for health management and disease monitoring. Over the past 10 years, advances in nanotechnology, microfluidics, and biomarker research have spurred the development of powerful miniaturized diagnostic systems for whole blood testing toward the goal of disease monitoring and treatment. Among the techniques employed for whole-blood diagnostics, electrochemical biosensors, as known to be rapid, sensitive, capable of miniaturization, reagentless and washing free, become a class of emerging technology to achieve the target detection specifically and directly in complex media, e.g., whole blood or even in the living body. Here we are aiming to provide a comprehensive review to summarize advances over the past decade in the development of electrochemical sensors for whole blood analysis. Further, we address the remaining challenges and opportunities to integrate electrochemical sensing platforms.
Collapse
Affiliation(s)
- Shaoguang Li
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Hongyuan Zhang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Man Zhu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Zhujun Kuang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Xun Li
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Fan Xu
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Siyuan Miao
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Zishuo Zhang
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Xiaoding Lou
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Hui Li
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| | - Fan Xia
- State Key Laboratory of Biogeology and Environmental Geology, Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
| |
Collapse
|
34
|
Cornish N, Haycock P, Brenner H, Figueiredo JC, Galesloot T, Grant RC, InterLymph consortium, INVENT-MVP consortium, Johansson M, Mariosa D, McKay J, Pai R, Pellatt AJ, Samadder NJ, Shi J, Thibord F, Trégouët DA, Voegele C, Thirlwell C, Mumford A, Langdon R. Causal relationships between risk of venous thromboembolism and 18 cancers: a bidirectional Mendelian randomisation analysis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.16.23289792. [PMID: 37292802 PMCID: PMC10246038 DOI: 10.1101/2023.05.16.23289792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background People with cancer experience high rates of venous thromboembolism (VTE). Additionally, risk of subsequent cancer is increased in people experiencing their first VTE. The causal mechanisms underlying this association are not completely understood, and it is unknown whether VTE is itself a risk factor for cancer. Methods We used data from large genome-wide association study meta-analyses to perform bi-directional Mendelian randomisation analyses to estimate causal associations between genetically-proxied lifetime risk of VTE and risk of 18 different cancers. Results We found no conclusive evidence that genetically-proxied lifetime risk of VTE was causally associated with an increased incidence of cancer, or vice-versa. We observed an association between VTE and pancreatic cancer risk (odds ratio for pancreatic cancer 1.23 (95% confidence interval 1.08 - 1.40) per log-odds increase in risk of VTE, P = 0.002). However, sensitivity analyses revealed this association was predominantly driven by a variant proxying non-O blood group, with inadequate evidence from Mendelian randomisation to suggest a causal relationship. Conclusions These findings do not support the hypothesis that genetically-proxied lifetime risk of VTE is a cause of cancer. Existing observational epidemiological associations between VTE and cancer are therefore more likely to be driven by pathophysiological changes which occur in the setting of active cancer and anti-cancer treatments. Further work is required to explore and synthesise evidence for these mechanisms.
Collapse
Affiliation(s)
- Naomi Cornish
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
- University of Exeter Medical School, University of Exeter, Exeter, UK
| | - Philip Haycock
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jane C. Figueiredo
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles CA
| | - Tessel Galesloot
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert C Grant
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | | | - Mattias Johansson
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Daniela Mariosa
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James McKay
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Rish Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Arizona, Scottsdale, USA
| | - Andrew J Pellatt
- Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Florian Thibord
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung and Blood Institute, Framingham, MA, USA
| | | | - Catherine Voegele
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | | | - Andrew Mumford
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Ryan Langdon
- Medical Research Council Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| |
Collapse
|
35
|
Xu S, Zhang XP, Zhao GD, Zou WB, Zhao ZM, Liu Q, Hu MG, Liu R. Derivation and validation of a preoperative prognostic model for resectable pancreatic ductal adenocarcinoma. Hepatobiliary Pancreat Dis Int 2023; 22:160-168. [PMID: 36171167 DOI: 10.1016/j.hbpd.2022.09.009] [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: 08/19/2021] [Accepted: 09/07/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND The prognosis of patients with pancreatic ductal adenocarcinoma (PDAC) remains poor even after radical pancreaticoduodenectomy (PD). The study aimed to develop and validate a novel preoperative prognostic model to accurately predict the long-term survival of patients with PDAC. METHODS Patients with PDAC of pancreatic head from Chinese PLA General Hospital were included. The preoperative PDAC model with contour plots was developed using a non-linear model in the training cohort and then tested in the validation cohort. RESULTS Of 421 patients who met the inclusion criteria, 280 were in the training cohort and 141 in the validation cohort. Contour plots for preoperative PDAC model were established to visually predict the survival probabilities of these patients, based on preoperative carbohydrate antigen 19-9, preoperative fibrinogen to albumin ratio and pain symptoms. This model stratified patients into low- and high-risk groups with distinctly different long-term survival in the training cohort [median overall survival (OS) 32.1 vs. 17.5 months; median recurrence-free survival (RFS) 19.3 vs. 10.0 months, both P < 0.001] and the validation cohort (median OS 28.3 vs. 19.0 months; median RFS 17.5 vs. 11.2 months, both P < 0.001). Time-dependent receiver operating characteristic and decision curve analyses revealed that the model provided higher diagnostic accuracy and superior net benefit compared to other staging systems. CONCLUSIONS This study constructed and validated a novel preoperative prognostic model that can accurately and conveniently predict the long-term survival of patients with resectable PDAC of pancreatic head. Besides, the model can screen high-risk patients with poor prognosis, which may provide references for personal treatment strategies in the future.
Collapse
Affiliation(s)
- Shuai Xu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China; Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Xiu-Ping Zhang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Guo-Dong Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Wen-Bo Zou
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhi-Ming Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Qu Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Ming-Gen Hu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Rong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
36
|
Li N, Zhang Y, Qu W, Zhang C, Ding Z, Wang L, Cui B. Analysis of systemic inflammatory and coagulation biomarkers in advanced cervical cancer: Prognostic and predictive significance. Int J Biol Markers 2023:3936155231163599. [PMID: 36927209 DOI: 10.1177/03936155231163599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
OBJECTIVE Peripheral systemic inflammatory, nutritional, and coagulation biomarkers have prognostic and predictive value in various malignancies. We evaluated the prognostic and predictive roles of systemic inflammatory, nutritional, and coagulation biomarkers in the circulating blood of patients with advanced cervical cancer. METHODS A retrospective study of 795 patients with cervical cancer who received concurrent chemoradiation therapy was performed. Overall survival was evaluated by the Kaplan-Meier estimator. Univariate and multivariate Cox regression models were used to determine prognostic factors associated with overall survival. RESULTS The median follow-up time was 76 months. In the univariate analysis, overall survival showed positive prognostic value in patients with a platelet-to-lymphocyte ratio (PLR) <164.29 (P = 0.010), and a plasma fibrinogen (FIB) level <4 g/L(P = 0.012). In the multivariate analysis, the PLR (P = 0.036), and FIB level (P = 0.047) maintained their significance for overall survival. Therefore, the PLR and FIB levels are independent prognostic factors in patients with advanced cervical cancer. CONCLUSIONS Systemic inflammatory and coagulation biomarkers could help to understand survival differences in the clinical treatment of advanced cervical cancer. The PLR and FIB levels are independent prognostic factors of poor survival in patients with advanced cervical cancer.
Collapse
Affiliation(s)
- Ningfeng Li
- Department of Gynecology, 117907Weifang People's Hospital, Weifang, China
| | - Yan Zhang
- Department of Gynecology, 117907Weifang People's Hospital, Weifang, China
| | - Wenjie Qu
- Department of Obstetrics and Gynecology, 91623Qilu Hospital of Shandong University, Jinan, China
| | - Chao Zhang
- Department of Emergency, 235960The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhaoxia Ding
- Department of Gynecology, 235960The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Academy of Medical Science, 71107Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Baoxia Cui
- Department of Obstetrics and Gynecology, 91623Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
37
|
Dobson DA, Holle LA, Lin FC, Huffman JE, Luyendyk JP, Flick MJ, Smith NL, de Vries PS, Morrison AC, Wolberg AS. Novel genetic regulators of fibrinogen synthesis identified by an in vitro experimental platform. J Thromb Haemost 2023; 21:522-533. [PMID: 36696182 PMCID: PMC10111212 DOI: 10.1016/j.jtha.2022.10.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/06/2022] [Accepted: 10/26/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Fibrinogen has an established, essential role in both coagulation and inflammatory pathways, and these processes are deeply intertwined in the development of thrombotic and atherosclerotic diseases. Previous studies aimed to better understand the (patho) physiological actions of fibrinogen by characterizing the genomic contribution to circulating fibrinogen levels. OBJECTIVES Establish an in vitro approach to define functional roles between genes within these loci and fibrinogen synthesis. METHODS Candidate genes were selected on the basis of their proximity to genetic variants associated with fibrinogen levels and expression in hepatocytes and HepG2 cells. HepG2 cells were transfected with small interfering RNAs targeting candidate genes and cultured in the absence or presence of the proinflammatory cytokine interleukin-6. Effects on fibrinogen protein production, gene expression, and cell growth were assessed by immunoblotting, real-time polymerase chain reaction, and cell counts, respectively. RESULTS HepG2 cells secreted fibrinogen, and stimulation with interleukin-6 increased fibrinogen production by 3.4 ± 1.2 fold. In the absence of interleukin-6, small interfering RNA knockdown of FGA, IL6R, or EEPD1 decreased fibrinogen production, and knockdown of LEPR, PDIA5, PLEC, SHANK3, or CPS1 increased production. In the presence of interleukin-6, knockdown of FGA, IL6R, or ATXN2L decreased fibrinogen production. Knockdown of FGA, IL6R, EEPD1, LEPR, PDIA5, PLEC, or CPS1 altered transcription of one or more fibrinogen genes. Knocking down ATXN2L suppressed inducible but not basal fibrinogen production via a post-transcriptional mechanism. CONCLUSIONS We established an in vitro platform to define the impact of select gene products on fibrinogen production. Genes identified in our screen may reveal cellular mechanisms that drive fibrinogen production as well as fibrin(ogen)-mediated (patho)physiological mechanisms.
Collapse
Affiliation(s)
- Dre'Von A Dobson
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, NC, USA
| | - Lori A Holle
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, NC, USA
| | - Feng-Chang Lin
- Department of Biostatistics and North Carolina Translational and Clinical Sciences Institute, University of North Carolina at Chapel Hill, NC, USA
| | | | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, USA
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, NC, USA
| | - Nicholas L Smith
- Department of Epidemiology, University of Washington, Seattle WA, USA; Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA, USA; Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle WA, USA; Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paul S de Vries
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA, USA
| | - Alanna C Morrison
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle WA, USA
| | - Alisa S Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, NC, USA.
| |
Collapse
|
38
|
Hu P, Wang W, He C. Fibrinogen-to-Lymphocyte Ratio Was an Independent Predictor of Lymph Node Metastasis in Patients with Clinically Node-Negative Advanced-Stage Gastric Cancer. Int J Gen Med 2023; 16:1345-1354. [PMID: 37089136 PMCID: PMC10120823 DOI: 10.2147/ijgm.s407833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023] Open
Abstract
Purpose Various hematological indicators have been reported to predict lymph node metastasis (LNM) in gastric cancer (GC) patients, but the relationship between FLR and LNM has not been studied. Therefore, the aim of this study was to evaluate the role of preoperative fibrinogen-to-lymphocyte ratio (FLR) in predicting LNM in patients with clinically node-negative (cN0) advanced gastric cancer (AGC). Patients and Methods We retrospectively reviewed 571 eligible patients with primary AGC adenocarcinoma who underwent radical gastrectomy (discovery cohort). Patients were divided into high and low FLR groups according to the optimal cutoff value determined by Youden index. FLR is an independent predictor of LNM determined by logistic regression and validated in the validation cohort of 207 patients. Receiver operating characteristic (ROC) curve analysis was used to evaluate the predictive value of FLR for LNM. The nonlinear relationship between FLR and LNM risk was assessed using restricted cubic spline. Sensitivity analyses were performed according to FLR quartiles to further assess the robustness of the results. The nomogram was built based on FLR and clinicopathological characteristics, and was evaluated by calibration curves, ROC curve analysis and decision curve analysis. Results In the discovery cohort, the area under the curve (AUC) value for FLR to predict LNM was 0.592. There is a linear relationship between the FLR value and the risk of LNM, and the risk of LNM increased with FLR value. High FLR level is an independent risk factor for LNM, and the results of sensitivity analysis robust this finding. The nomogram for individual risk assessment performed well. Furthermore, we verified the FLR was an independent predictor of LNM in the validation cohort. Conclusion FLR was an independent predictor of LNM in patients with cN0 AGC.
Collapse
Affiliation(s)
- Pei Hu
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, People’s Republic of China
| | - Wei Wang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, People’s Republic of China
- Correspondence: Wei Wang, Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, No. 2, Zheshan West Road, Wuhu, Anhui, 241000, People’s Republic of China, Tel +86-0553-5739316, Email
| | - Chiyi He
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui, People’s Republic of China
| |
Collapse
|
39
|
Wang J, Huang D, Wang Y, Yuan Q, Chen X, Cheng Y. Pretreatment plasma fibrinogen and serum albumin levels predict therapeutic efficacy of concurrent radiochemotherapy for esophageal squamous cell cancer. Front Oncol 2022; 12:1021214. [PMCID: PMC9610838 DOI: 10.3389/fonc.2022.1021214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeEvidence implies that plasma fibrinogen and serum albumin level (FA score) based on plasma fibrinogen and serum albumin is related to cancer prognosis. However, the association between the FA score and therapeutic efficacy of concurrent radiochemotherapy in esophageal squamous cell carcinoma (ESCC) has not yet been evaluated. This study aimed to assess the role of pretreatment FA score in predicting the therapeutic efficacy of concurrent radiochemotherapy for patients with esophageal squamous cell cancer.MethodsThis retrospective study evaluated 154 patients with ESCC who underwent concurrent radiochemotherapy. Receiver operating characteristic curve (ROC) analysis was used to determine the appropriate cut-off values, and multivariate analysis and Kaplan-Meier curve were used to evaluate prognosis.ResultsFA score was significantly associated with the N stage and M stage (P = 0.015 and 0.042, respectively). Chi-square analysis/Fisher’s exact tests revealed a correlation between the FA score and curative effect (P < 0.001), and higher FA score was associated with poorer treatment effect. Multivariate analysis indicated that FA score (P < 0.001) was predictor of overall survival (OS). Kaplan-Meier curve demonstrated that pretreatment FA score was significantly associated with the OS of ESCC: Patient with higher FA score has lower median OS.ConclusionsThe FA score is a reliable prognostic predictor that could assess the curative effect and OS benefit of concurrent radiochemotherapy in patients with ESCC.
Collapse
Affiliation(s)
- Jijin Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Di Huang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Wang
- Department of Oncology, Linyi People’s Hospital, Dezhou, China
| | - Qianqian Yuan
- Department of Oncology, Tengzhou Central People’s Hospital, Zaozhuang, China
| | - Xue Chen
- Department of Minimally Invasion Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Yufeng Cheng, ; Xue Chen,
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Yufeng Cheng, ; Xue Chen,
| |
Collapse
|
40
|
Li B, Deng H, Lei B, Chen L, Zhang X, Sha D. The prognostic value of fibrinogen to albumin ratio in malignant tumor patients: A meta-analysis. Front Oncol 2022; 12:985377. [PMID: 36249067 PMCID: PMC9556778 DOI: 10.3389/fonc.2022.985377] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundRecent studies have shown that the fibrinogen to albumin ratio (FAR) is closely related to the prognosis of various cancers. The aim of this systematic review and meta-analysis was to investigate the prognostic value of FAR in malignancies based on the available evidence.MethodTo systematically search the Cochrane Library, Embase, PubMed, Google Scholar, Baidu scholars, CNKI and VIP databases for relevant studies published before April 1, 2022, and to evaluate the fibrinogen-to-albumin ratio (FAR) and survival of patients with malignant tumors through a meta-analysis relationship between the results. Results. This meta-analysis included 19 eligible studies involving 5926 cancer patients. We found that high FAR was associated with poor overall survival (HR=2.25, 95%CI 1.86-2.74, p<0.001), recurrence-free survival (HR=2.29, 95%CI 1.91-2.76, P<0.001), progression-free survival (HR: 2.10, 95%CI 1.58-2.79, p<0.001), disease-free survival (HR=1.52, 95%CI 1.17-1.96, p=0.001), and time to recurrence (HR: 1.555, 95%CI 1.031-2.346, P=0.035) was significantly correlated.ConclusionsHigh FAR is significantly associated with poor clinical outcomes in cancer, suggesting that it may be an important predictor of prognosis in patients with malignancies.
Collapse
Affiliation(s)
- Baibei Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huachu Deng
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Biao Lei
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Leijie Chen
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xinyuan Zhang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Dingran Sha
- Department of Urology Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Dingran Sha,
| |
Collapse
|
41
|
Zhang XP, Gao YX, Xu S, Zhao GD, Hu MG, Tan XL, Zhao ZM, Liu R. A novel online calculator to predict early recurrence and long-term survival of patients with resectable pancreatic ductal adenocarcinoma after pancreaticoduodenectomy: A multicenter study. Int J Surg 2022; 106:106891. [PMID: 36165934 DOI: 10.1016/j.ijsu.2022.106891] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/16/2022] [Accepted: 08/28/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is prone to relapse even after radical pancreaticoduodenectomy (PD) (including robotic, laparoscopic and open approach). This study aimed to develop an online nomogram calculator to predict early recurrence (ER) (within one year after surgery) and long-term survival in patients with PDAC. METHODS Patients with PDAC after radical PD were included. Univariate and multivariate logistic regression analysis was used to identify independent risk factors. An online nomogram calculator was developed based on independent risk factors in the training cohort and then tested in the internal and external validation cohorts. RESULTS Of the 569 patients who met the inclusion criteria, 310, 155, and 104 patients were in the training, internal and external validation cohorts, respectively. Multivariate analysis revealed that preoperative carbohydrate antigen19-9 (CA19-9) [Odds Ratio (OR) 1.002; 95% confidence interval (CI) 1.001-1.003; P = 0.001], fibrinogen/albumin (FAR) (OR 1.132; 95% CI 1.012-1.266; P = 0.029), N stage (OR 2.291; 95% CI 1.283-4.092; P = 0.005), and tumor differentiation (OR 3.321; 95% CI 1.278-8.631; P = 0.014) were independent risk factors for ER. Nomogram based on the above four factors achieved good C-statistics of 0.772, 0.767 and 0.765 in predicting ER in the training, internal and external validation cohorts, respectively. Time-dependent ROC analysis (timeROC) and decision curve analysis (DCA) revealed that the nomogram provided superior diagnostic capacity and net benefit compared with other staging systems. CONCLUSION This multi-center study developed and validated an online nomogram calculator that can predict ER and long-term survival in patients with PDAC with high degrees of stability and accuracy.
Collapse
Affiliation(s)
- Xiu-Ping Zhang
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuan-Xing Gao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Shuai Xu
- Department of Liver Transplantation and Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guo-Dong Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Ming-Gen Hu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiang-Long Tan
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhi-Ming Zhao
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.
| | - Rong Liu
- Faculty of Hepato-Biliary-Pancreatic Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.
| |
Collapse
|
42
|
Liang L, Liu F, Yang W, Yang W, Chen L, He Y, Liu Z, Zhang L, Zhang F, Cai F, Xu H, Lin M, Liu M, Pan Y, Liu Y, Hu Z, Chen H, He Z, Ke Y. Combined Mean Corpuscular Hemoglobin, Fibrinogen, and Albumin (MF-A) Is a Novel Prognostic Marker in Patients with Resectable Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2022; 29:5626-5633. [PMID: 35181817 DOI: 10.1245/s10434-022-11415-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND The aim was to systematically select blood markers routinely tested in clinical settings, which are independently associated with overall survival (OS) and are able to stratify prognosis of esophageal squamous cell carcinoma (ESCC) patients undergoing esophagectomy. METHODS We selected optimal blood markers for prognostic stratification from 60 candidates in a clinical cohort of 1819 consecutive patients with resectable ESCC in China. Selection was carried out using two-step multivariable Cox proportional hazards regression adjusted for multifaceted confounders. A composite index was developed by multiplying risk factors and dividing them by protective factors. RESULTS With a median follow-up of 48.07 months, 641 deaths occurred in the 1819 patients and the 5-year OS was 56.30%. Two risk factors (mean corpuscular hemoglobin, fibrinogen) and a protective factor (albumin), all dichotomized and assigned values 1 and 2, were used to construct the composite index marker "MF-A". Three risk groups were created based on the MF-A score including low- (0.5), moderate- (1), and high-risk groups (2 and 4). Compared with patients in the low-risk group (1184/1778, 66.59%), those in the moderate- (488, 27.45%), and high-risk (106, 5.96%) groups were at elevated risk of death (adjusted HR: 1.32, 95% CI: 1.11-1.57; adjusted HR: 2.08, 95% CI: 1.56-2.75; Ptrend < 10-7). Within each TNM stage grouping, OS also trended to be significantly worse as the MF-A score increased. CONCLUSIONS "MF-A" is a novel independent predictor which may be used to estimate and stratify prognosis for ESCC patients undergoing esophagectomy.
Collapse
Affiliation(s)
- Linlin Liang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Fangfang Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Wenlei Yang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Wei Yang
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Lei Chen
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Yu He
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene & Tropical Medicine, London, UK
| | - Zhen Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Liqun Zhang
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Fan Zhang
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Fen Cai
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Huawen Xu
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Miaoping Lin
- Cancer Hospital of Shantou University Medical College, Shantou City, People's Republic of China
| | - Mengfei Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yaqi Pan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Ying Liu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhe Hu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Huanyu Chen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Zhonghu He
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| | - Yang Ke
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Genetics, Peking University Cancer Hospital & Institute, Beijing, People's Republic of China
| |
Collapse
|
43
|
Morris K, Schnoor B, Papa AL. Platelet cancer cell interplay as a new therapeutic target. Biochim Biophys Acta Rev Cancer 2022; 1877:188770. [DOI: 10.1016/j.bbcan.2022.188770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 10/16/2022]
|
44
|
Abstract
Tissue factor (TF), an initiator of extrinsic coagulation pathway, is positively correlated with venous thromboembolism (VTE) of tumor patients. Beyond thrombosis, TF plays a vital role in tumor progression. TF is highly expressed in cancer tissues and circulating tumor cell (CTC), and activates factor VIIa (FVIIa), which increases tumor cells proliferation, angiogenesis, epithelial-mesenchymal transition (EMT) and cancer stem cells(CSCs) activity. Furthermore, TF and TF-positive microvesicles (TF+MVs) activate the coagulation system to promote the clots formation with non-tumor cell components (e.g., platelets, leukocytes, fibrin), which makes tumor cells adhere to clots to form CTC clusters. Then, tumor cells utilize clots to cause its reducing fluid shear stress (FSS), anoikis resistance, immune escape, adhesion, extravasation and colonization. Herein, we review in detail that how TF signaling promotes tumor metastasis, and how TF-targeted therapeutic strategies are being in the preclinical and clinical trials.
Collapse
|
45
|
Mai RY, Bai T, Luo XL, Wu GB. Preoperative fibrinogen-to-albumin ratio predicts the prognosis of patients with hepatocellular carcinoma subjected to hepatectomy. BMC Gastroenterol 2022; 22:261. [PMID: 35606690 PMCID: PMC9128092 DOI: 10.1186/s12876-022-02328-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Background Systemic inflammatory response (SIR) plays a crucial role in every step of tumorigenesis and development. More recently, the fibrinogen-to-albumin ratio (FAR), an inflammation-based model, was suggested as a prognostic maker for various cancer patients. This research aimed to estimate the prognostic abilities of FAR, neutrophil–lymphocyte ratio (NLR), monocyte-lymphocyte ratio (MLR), platelet– lymphocyte ratio (PLR), and systemic immune–inflammation index (SII) in patients with hepatocellular carcinoma (HCC) subjected to curative hepatectomy. Methods A total of 1,502 cases who underwent hepatectomy for HCC were included. The predictive performances of FAR, NLR, MLR, PLR and SII were assessed with regards to overall survival (OS) and disease-free survival (DFS). The area under the time-dependent receiver operating characteristic curve was used to compare prognostic performances. Results Data revealed that FAR had higher predictive accuracy than other inflammation-based models and alpha-fetoprotein (AFP) in assessing OS and DFS. Indeed, the OS and DFS of patients with high FAR (> 8.9), differentiated by the optimal cut-off value of FAR, were remarkably reduced (p < 0.05 for OS and DFS). Multivariate Cox regression analyses identified that AFP, FAR, clinically significant portal hypertension, tumor size, Barcelona Clinical Liver Cancer staging system, major resection and blood loss were independent indicators for predicting OS and DFS. Furthermore, these patients could be classified according to their FAR into significantly different subgroups, regardless of AFP levels (p < 0.05 for DFS and OS). Similar results were obtained in other inflammation-based prognostic models. Conclusions Compared with NLR, MLR, PLR, SII and AFP, FAR showed significant advantages in predicting survival of HCC patients subjected to liver resection. Supplementary Information The online version contains supplementary material available at 10.1186/s12876-022-02328-4.
Collapse
Affiliation(s)
- Rong-Yun Mai
- Department of Hepatobilliary and Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.,Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Tao Bai
- Department of Hepatobilliary and Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China
| | - Xiao-Ling Luo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| | - Guo-Bin Wu
- Department of Hepatobilliary and Pancreatic Surgery, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
46
|
Cai L, Tu M, Yin X, Zhang S, Zhuang W, Xia Y, Zhang Y, Zhang L, Yu L, Chi L, Huang Y. Combination of Serum CST4 and DR-70 Contributes to Early Diagnosis of Colorectal Cancer. Clin Chim Acta 2022; 531:318-324. [DOI: 10.1016/j.cca.2022.04.1000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/26/2022] [Indexed: 11/26/2022]
|
47
|
Sano J, Matsuda S, Kawakubo H, Takemura R, Okui J, Irino T, Fukuda K, Nakamura R, Kitagawa Y. Exposure to a Postoperative Hypercoagulable State Predicts Poor Prognosis After Transthoracic Esophagectomy in Patients with Esophageal Cancer. Ann Surg Oncol 2022; 29:10.1245/s10434-022-11591-4. [PMID: 35347519 DOI: 10.1245/s10434-022-11591-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/21/2022] [Indexed: 02/21/2024]
Abstract
PURPOSE The contribution of postoperative coagulation-fibrinolysis status to prognosis is yet to be fully investigated. Thus, in this study, we aimed to elucidate the relationship between postoperative hypercoagulable state (PHS) after transthoracic esophagectomy and long-term outcome in patients with esophageal cancer. METHODS Patients with esophageal cancer who underwent transthoracic esophagectomy were selected from a prospectively maintained database. Based on the trend of postoperative plasma fibrin-fibrinogen degradation product (FDP) levels, patients with PHS were identified. The prognostic significance of PHS was evaluated via multivariate analysis using the Cox regression model. RESULTS Based on the plasma FDP levels of 172 patients that reached a plateau between POD5 and POD7, we calculated the mean FDP value of POD5, 6, and 7, setting a median value as a cutoff. Consequently, 87 patients were classified as PHS. The overall survival (OS) in the PHS group was determined to be significantly lower than in the non-PHS group (5-year OS; 68% and 80%, p = 0.012). Recurrence-free survival (RFS) in the PHS group was significantly lower than in the non-PHS group (5-year RFS; 60% and 79%, p = 0.017). Using the pathological stage as a covariate in the multivariate analysis, PHS was an independent prognostic factor of OS [hazard ratio (HR) 2.517, p = 0.009] and RFS (HR 1.905, p = 0.041). CONCLUSIONS PHS was found to be an independent negative prognostic factor in patients with esophageal cancer. Possible improvement of the oncological outcome by early postoperative intervention with anticoagulants should be explored in clinical trials.
Collapse
Affiliation(s)
- Junichi Sano
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Hirofumi Kawakubo
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Ryo Takemura
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Jun Okui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyuki Irino
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazumasa Fukuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Rieko Nakamura
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
48
|
Juang LJ, Hur WS, Silva LM, Strilchuk AW, Francisco B, Leung J, Robertson MK, Groeneveld DJ, La Prairie B, Chun EM, Cap AP, Luyendyk JP, Palumbo JS, Cullis PR, Bugge TH, Flick MJ, Kastrup CJ. Suppression of fibrin(ogen)-driven pathologies in disease models through controlled knockdown by lipid nanoparticle delivery of siRNA. Blood 2022; 139:1302-1311. [PMID: 34958662 PMCID: PMC8900269 DOI: 10.1182/blood.2021014559] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 12/14/2021] [Indexed: 11/20/2022] Open
Abstract
Fibrinogen plays a pathologic role in multiple diseases. It contributes to thrombosis and modifies inflammatory and immune responses, supported by studies in mice expressing fibrinogen variants with altered function or with a germline fibrinogen deficiency. However, therapeutic strategies to safely and effectively tailor plasma fibrinogen concentration are lacking. Here, we developed a strategy to tune fibrinogen expression by administering lipid nanoparticle (LNP)-encapsulated small interfering RNA (siRNA) targeting the fibrinogen α chain (siFga). Three distinct LNP-siFga reagents reduced both hepatic Fga messenger RNA and fibrinogen levels in platelets and plasma, with plasma levels decreased to 42%, 16%, and 4% of normal within 1 week of administration. Using the most potent siFga, circulating fibrinogen was controllably decreased to 32%, 14%, and 5% of baseline with 0.5, 1.0, and 2.0 mg/kg doses, respectively. Whole blood from mice treated with siFga formed clots with significantly decreased clot strength ex vivo, but siFga treatment did not compromise hemostasis following saphenous vein puncture or tail transection. In an endotoxemia model, siFga suppressed the acute phase response and decreased plasma fibrinogen, D-dimer, and proinflammatory cytokine levels. In a sterile peritonitis model, siFga restored normal macrophage migration in plasminogen-deficient mice. Finally, treatment of mice with siFga decreased the metastatic potential of tumor cells in a manner comparable to that observed in fibrinogen-deficient mice. The results indicate that siFga causes robust and controllable depletion of fibrinogen and provides the proof-of-concept that this strategy can modulate the pleiotropic effects of fibrinogen in relevant disease models.
Collapse
Affiliation(s)
- Lih Jiin Juang
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Woosuk S Hur
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lakmali M Silva
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Amy W Strilchuk
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Brenton Francisco
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Jerry Leung
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Madelaine K Robertson
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Dafna J Groeneveld
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Bridget La Prairie
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth M Chun
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Andrew P Cap
- The United States Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX
- Department of Medicine, Uniformed Services University, Bethesda, MD
| | - James P Luyendyk
- Department of Pathobiology and Diagnostic Investigation, Institute for Integrative Toxicology, Michigan State University, East Lansing, MI
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Matthew J Flick
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Christian J Kastrup
- Michael Smith Laboratories
- Centre for Blood Research, and
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Blood Research Institute, Versiti, Milwaukee, WI; and
- Department of Surgery, Department of Biochemistry, Department of Biomedical Engineering, and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
49
|
Xie H, Yuan G, Liu M, Huang S, Wei L, Tang S, Gan J. Pretreatment Albumin-to-Fibrinogen Ratio is a Promising Biomarker for Predicting Postoperative Clinical Outcomes in Patients with Colorectal Cancer. Nutr Cancer 2022; 74:2896-2909. [PMID: 35193433 DOI: 10.1080/01635581.2022.2042572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE To evaluate the prognostic value of pretreatment albumin-to-fibrinogen ratio (AFR) in colorectal cancer (CRC). METHODS This retrospective study included 657 CRC patients who underwent surgical resection in 2012-2014. Kaplan-Meier survival curve and Cox proportional hazards model were used to determine independent predictors. Receiver operating characteristic curve analysis was used to assess and compare the ability of indicators to predict survival. RESULTS The optimal cutoff value of AFR was 8.3. Compared with high AFR group, low AFR group had shorter progression-free survival (PFS) (65.32% vs 52.28%, p < 0.001) and overall survival (OS) (67.47% vs 56.14%, p = 0.001). In the stratified analysis of TNM stage, AFR had good prognostic discrimination for early- and advanced-stage patients. Multivariate analysis suggested that AFR was an independent prognostic factor of PFS [hazard ratio (HR) = 1.385, 95% confidence interval (CI) = 1.043-1.839, p = 0.024) and OS (HR = 1.342, 95% CI = 1.022-1.763, p = 0.034) for CRC patients. AFR had better prognostic prediction ability than other inflammation-related markers. The AFR-based nomograms had good predictive capabilities. CONCLUSIONS Pretreatment AFR is an independent prognostic factor for CRC patients undergoing surgical resection and is superior to other established inflammation-related markers.
Collapse
Affiliation(s)
- Hailun Xie
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Guanghui Yuan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Mingxiang Liu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Shizhen Huang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Lishuang Wei
- Department of Respiratory Medicine, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Shuangyi Tang
- Department of Pharmacy, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| | - Jialiang Gan
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, P.R. China
| |
Collapse
|
50
|
Chen W, Shan B, Zhou S, Yang H, Ye S. Fibrinogen/albumin ratio as a promising predictor of platinum response and survival in ovarian clear cell carcinoma. BMC Cancer 2022; 22:92. [PMID: 35062908 PMCID: PMC8780809 DOI: 10.1186/s12885-022-09204-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
Background This study aims to evaluate the role of the fibrinogen/albumin ratio (FAR) in predicting platinum resistance and survival outcomes of patients with ovarian clear cell carcinoma (OCCC). Methods Coagulation function and D-dimer, serum albumin, CA125 and HE4 levels were measured before surgery in OCCC patients undergoing initial surgery in our institution. FAR was calculated as fibrinogen/albumin level. The correlation between these indicators and clinicopathological features, platinum response, and survival outcomes was further analyzed. The Kaplan-Meier method and multivariable Cox regression model were used to assess the effects of FAR on progression-free survival (PFS) and overall survival (OS). Results Advanced stage patients accounted for 42.1% of the 114 participants. Optimal cytoreductive surgery was achieved in 105 patients, and the complete resection rate was 78.1%. FAR was associated with tumor stage, residual tumor and platinum response. A receiver operating characteristic curve for predicting platinum response showed that the optimal cutoff point of the FAR was 12%. The sensitivity was 73.3% and the specificity was 68.2%. In multivariate analysis, FAR ≥12% (HR = 4.963, P = 0.002) was an independent risk factor for platinum resistance. In addition, FAR and D-dimer proved to be independent negative factors for outcomes including both PFS and OS. The median follow-up time was 52 months. A high FAR (≥ 12%) showed a stronger correlation with poor OS and PFS in the subgroup analysis of advanced and completely resected patients. Conclusions The FAR might be a potential preoperative biochemical marker for predicting treatment response and oncological outcomes in OCCC patients.
Collapse
|