51
|
Yang WY, Izzi B, Bress AP, Thijs L, Citterio L, Wei FF, Salvi E, Delli Carpini S, Manunta P, Cusi D, Hoylaerts MF, Luttun A, Verhamme P, Hardikar S, Nawrot TS, Staessen JA, Zhang ZY. Association of colorectal cancer with genetic and epigenetic variation in PEAR1—A population-based cohort study. PLoS One 2022; 17:e0266481. [PMID: 35390065 PMCID: PMC8989234 DOI: 10.1371/journal.pone.0266481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
Platelet Endothelial Aggregation Receptor 1 (PEAR1) modulates angiogenesis and platelet contact-induced activation, which play a role in the pathogenesis of colorectal cancer. We therefore tested the association of incident colorectal cancer and genetic and epigenetic variability in PEAR1 among 2532 randomly recruited participants enrolled in the family-based Flemish Study on Environment, Genes and Health Outcomes (51.2% women; mean age 44.8 years). All underwent genotyping of rs12566888 located in intron 1 of the PEAR1 gene; in 926 participants, methylation at 16 CpG sites in the PEAR1 promoter was also assessed. Over 18.1 years (median), 49 colorectal cancers occurred, all in different pedigrees. While accounting for clustering of risk factors within families and adjusting for sex, age, body mass index, the total-to-HDL cholesterol ratio, serum creatinine, plasma glucose, smoking and drinking, use of antiplatelet and nonsteroidal anti-inflammatory drug, the hazard ratio of colorectal cancer contrasting minor-allele (T) carriers vs. major-allele (GG) homozygotes was 2.17 (95% confidence interval, 1.18–3.99; P = 0.013). Bootstrapped analyses, from which we randomly excluded from two to nine cancer cases, provided confirmatory results. In participants with methylation data, we applied partial least square discriminant analysis (PLS-DA) and identified two methylation sites associated with higher colorectal cancer risk and two with lower risk. In-silico analysis suggested that methylation of the PEAR1 promoter at these four sites might affect binding of transcription factors p53, PAX5, and E2F-1, thereby modulating gene expression. In conclusion, our findings suggest that genetic and epigenetic variation in PEAR1 modulates the risk of colorectal cancer in white Flemish. To what extent, environmental factors as exemplified by our methylation data, interact with genetic predisposition and modulate penetrance of colorectal cancer risk is unknown.
Collapse
Affiliation(s)
- Wen-Yi Yang
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Benedetta Izzi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, Pozzilli, Italy
| | - Adam P Bress
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, United States of America
| | - Lutgarde Thijs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lorena Citterio
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fang-Fei Wei
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Erika Salvi
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Simona Delli Carpini
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Manunta
- School of Nephrology, University Vita-Salute San Raffaele, Milan, Italy
| | | | | | - Aernout Luttun
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Peter Verhamme
- Center for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Sheetal Hardikar
- Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Tim S Nawrot
- Centre for Environmental Sciences, Hasselt University, Hasselt, Belgium
| | - Jan A Staessen
- Biomedical Science Group, University of Leuven, Leuven, Belgium
- Research Institute Association for the Promotion of Preventive Medicine, Mechelen, Belgium
| | - Zhen-Yu Zhang
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
52
|
Bulla SC, Badial PR, Bulla C. Canine Cancer Cells Activate Platelets via the Platelet P2Y12 Receptor. J Comp Pathol 2022; 192:41-49. [PMID: 35305713 DOI: 10.1016/j.jcpa.2022.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 11/09/2021] [Accepted: 01/13/2022] [Indexed: 11/16/2022]
Abstract
In addition to their well-known functions in haemostasis, anucleated platelets have a critical role in cancer biology. Many human and non-human cancer types can directly interact with and activate platelets, promoting cancer malignancy and progression. Although naturally occurring canine neoplastic diseases mimic the biologically complex conditions of human cancers more closely than laboratory-bred mice, studies evaluating the relationship between cancer cells and platelets in dogs are scarce, and the effects of tumour cells on platelets in these animals are unknown. To evaluate whether cancer cells could activate canine platelets, we assessed the response of platelet-rich plasma to cultured canine cancer cells using light transmittance aggregometry. Similar to human and murine cancer cell research, we demonstrated that both canine osteosarcoma and mammary carcinoma cells activated canine platelets in vitro, resulting in platelet aggregation. The degree of aggregation was most pronounced at a cancer cell to platelet ratio of 1:200 for most cell lines. Mechanistic studies revealed that the platelet adenosine diphosphate (ADP) receptor P2Y12 is essential for canine platelet aggregation induced by canine cancer. ADP receptor blockage on platelets inhibited >50% of cancer cell-induced maximum platelet aggregation in all cell lines evaluated. As in other species, our results suggest that canine cancers may activate canine platelets in vivo. This mechanism is likely relevant for the biology and progression of cancer in the dog.
Collapse
Affiliation(s)
- Sandra C Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Peres R Badial
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA
| | - Camilo Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, Mississippi, USA.
| |
Collapse
|
53
|
Giannakeas V. Trends in platelet count among cancer patients. Exp Hematol Oncol 2022; 11:16. [PMID: 35331331 PMCID: PMC8944120 DOI: 10.1186/s40164-022-00272-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/18/2022] [Indexed: 12/05/2022] Open
Abstract
An elevated platelet count has been associated with an increased incidence of cancer and poor survival for many cancer types. In this study, platelet levels were captured among cancer patients in the 2 years prior to and following a cancer diagnosis. I investigated if the trends in platelet count differ between patients that died or did not die from their cancer. For many cancer types, including colon, lung, ovary, and stomach, platelet counts rose as they approached the date of diagnosis. Patients that died from their cancer within 3 years of diagnosis had a higher peak platelet count than those who survived. Following diagnosis, platelet count was elevated among patients that died from their cancer as compared to patients who survived. An elevated platelet count could potentially indicate the presence of an occult cancer or be used as a prognostic measure for cancer-specific survival.
Collapse
Affiliation(s)
- Vasily Giannakeas
- Women's College Research Institute, Women's College Hospital, 76 Grenville Street, 6th Floor, Toronto, ON, M5S 1B2, Canada. .,Dalla Lana School of Public Health, University of Toronto, 155 College Street, Health Science Building, 6th Floor, Toronto, ON, Canada. .,ICES, Toronto, ON, Canada.
| |
Collapse
|
54
|
Ogris C, Castresana-Aguirre M, Sonnhammer ELL. PathwAX II: Network-based pathway analysis with interactive visualization of network crosstalk. Bioinformatics 2022; 38:2659-2660. [PMID: 35266519 PMCID: PMC9048662 DOI: 10.1093/bioinformatics/btac153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/03/2022] [Accepted: 03/09/2022] [Indexed: 11/28/2022] Open
Abstract
Motivation Pathway annotation tools are indispensable for the interpretation of a wide range of experiments in life sciences. Network-based algorithms have recently been developed which are more sensitive than traditional overlap-based algorithms, but there is still a lack of good online tools for network-based pathway analysis. Results We present PathwAX II—a pathway analysis web tool based on network crosstalk analysis using the BinoX algorithm. It offers several new features compared with the first version, including interactive graphical network visualization of the crosstalk between a query gene set and an enriched pathway, and the addition of Reactome pathways. Availability and implementation PathwAX II is available at http://pathwax.sbc.su.se. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Christoph Ogris
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 17121 Solna, Box, Sweden 1031.,Institute of Computational Biology, Helmholtz Center Munich, Neuherberg, Germany Ingolstädter Landstr. 1 85764
| | - Miguel Castresana-Aguirre
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 17121 Solna, Box, Sweden 1031
| | - Erik L L Sonnhammer
- Department of Biochemistry and Biophysics, Stockholm University, Science for Life Laboratory, 17121 Solna, Box, Sweden 1031
| |
Collapse
|
55
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
56
|
Rao C, Jin J, Lu J, Wang C, Wu Z, Zhu Z, Tu M, Su Z, Li Q. A Multielement Prognostic Nomogram Based on a Peripheral Blood Test, Conventional MRI and Clinical Factors for Glioblastoma. Front Neurol 2022; 13:822735. [PMID: 35250826 PMCID: PMC8893080 DOI: 10.3389/fneur.2022.822735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundGlioblastoma (GBM) is one of the most malignant types of tumors in the central nervous system, and the 5-year survival remains low. Several studies have shown that preoperative peripheral blood tests and preoperative conventional Magnetic Resonance Imaging (MRI) examinations affect the prognosis of GBM patients. Therefore, it is necessary to construct a risk score based on a preoperative peripheral blood test and conventional MRI and develop a multielement prognostic nomogram for GBM.MethodsThis study retrospectively analyzed 131 GBM patients. Determination of the association between peripheral blood test variables and conventional MRI variables and prognosis was performed by univariate Cox regression. The nomogram model, which was internally validated using a cohort of 56 GBM patients, was constructed by multivariate Cox regression. RNA sequencing data from Gene Expression Omnibus (GEO) and Chinese Glioma Genome Atlas (CGGA datasets were used to determine peripheral blood test-related genes based on GBM prognosis.ResultsThe constructed risk score included the neutrophil/lymphocyte ratio (NLR), lymphocyte/monocyte ratio (LMR), albumin/fibrinogen (AFR), platelet/lymphocyte ratio (PLR), and center point–to-ventricle distance (CPVD). A final nomogram was developed using factors associated with prognosis, including age, sex, the extent of tumor resection, IDH mutation status, radiotherapy status, chemotherapy status, and risk. The Area Under Curve (AUC) values of the receiver operating characteristic curve (ROC) curve were 0.876 (12-month ROC), 0.834 (24-month ROC) and 0.803 (36-month ROC) in the training set and 0.906 (12-month ROC), 0.800 (18-month ROC) and 0.776 (24-month ROC) in the validation set. In addition, vascular endothelial growth factor A (VEGFA) was closely associated with NLR and LMR and identified as the most central negative gene related to the immune microenvironment and influencing immune activities.ConclusionThe risk score was established as an independent predictor of GBM prognosis, and the nomogram model exhibit appropriate predictive power. In addition, VEGFA is the key peripheral blood test-related gene that is significantly associated with poor prognosis.
Collapse
|
57
|
Platelet Membrane: An Outstanding Factor in Cancer Metastasis. MEMBRANES 2022; 12:membranes12020182. [PMID: 35207103 PMCID: PMC8875259 DOI: 10.3390/membranes12020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/02/2022]
Abstract
In addition to being biological barriers where the internalization or release of biomolecules is decided, cell membranes are contact structures between the interior and exterior of the cell. Here, the processes of cell signaling mediated by receptors, ions, hormones, cytokines, enzymes, growth factors, extracellular matrix (ECM), and vesicles begin. They triggering several responses from the cell membrane that include rearranging its components according to the immediate needs of the cell, for example, in the membrane of platelets, the formation of filopodia and lamellipodia as a tissue repair response. In cancer, the cancer cells must adapt to the new tumor microenvironment (TME) and acquire capacities in the cell membrane to transform their shape, such as in the case of epithelial−mesenchymal transition (EMT) in the metastatic process. The cancer cells must also attract allies in this challenging process, such as platelets, fibroblasts associated with cancer (CAF), stromal cells, adipocytes, and the extracellular matrix itself, which limits tumor growth. The platelets are enucleated cells with fairly interesting growth factors, proangiogenic factors, cytokines, mRNA, and proteins, which support the development of a tumor microenvironment and support the metastatic process. This review will discuss the different actions that platelet membranes and cancer cell membranes carry out during their relationship in the tumor microenvironment and metastasis.
Collapse
|
58
|
Giannakeas V, Kotsopoulos J, Brooks JD, Cheung MC, Rosella L, Lipscombe L, Akbari MR, Austin PC, Narod SA. Platelet Count and Survival after Cancer. Cancers (Basel) 2022; 14:cancers14030549. [PMID: 35158817 PMCID: PMC8833779 DOI: 10.3390/cancers14030549] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Platelets are cellular fragments circulating in the blood that are responsible for clotting. Previous research has shown that cancer patients with an abnormally high platelet count (thrombocytosis) have elevated rates of death from cancer. We aimed to investigate to what extent platelet counts are associated with survival after cancer. We followed a large provincial cohort of cancer patients with a platelet count recorded at the time of their diagnosis. We categorized patients according to platelet count (low, medium, high). Cancer patients in the ‘high’ platelet count category had the highest rate of cancer death, and cancer patients in the ‘low’ platelet count category had the lowest rate of cancer death. Platelet count may be used to predict survival in cancer patients. Abstract Thrombocytosis is associated with cancer progression and death for many cancer types. It is unclear if platelet count is also associated with cancer survival. We conducted a cohort study of 112,231 adults in Ontario with a diagnosis of cancer between January 2007 and December 2016. We included patients who had a complete blood count (CBC) completed in the 30 days prior to their cancer diagnosis. Subjects were assigned to one of three categories according to platelet count: low (≤25th percentile), medium (>25 to <75th percentile), and high (≥75th percentile). Study subjects were followed from the date of their cancer diagnosis for cancer-specific death. Of the 112,231 eligible cancer patients in the cohort study, 40,329 (35.9%) died from their cancer in the follow-up period. Relative to those with a medium platelet count, the rate of cancer-specific death was higher among individuals with a high platelet count (HR 1.52; 95%CI 1.48–1.55) and was lower among individuals with a low platelet count (HR 0.91; 95%CI 0.88–0.93). A high platelet count was associated with poor survival for many cancer types. Platelet count could potentially be used as a risk stratification measure for cancer patients.
Collapse
Affiliation(s)
- Vasily Giannakeas
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON M5S 1B2, Canada; (V.G.); (J.K.); (L.L.); (M.R.A.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
- ICES, Toronto, ON M4N 3M5, Canada; (M.C.C.); (P.C.A.)
| | - Joanne Kotsopoulos
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON M5S 1B2, Canada; (V.G.); (J.K.); (L.L.); (M.R.A.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
| | - Jennifer D. Brooks
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
| | - Matthew C. Cheung
- ICES, Toronto, ON M4N 3M5, Canada; (M.C.C.); (P.C.A.)
- Division of Medical Oncology and Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
| | - Laura Rosella
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
| | - Lorraine Lipscombe
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON M5S 1B2, Canada; (V.G.); (J.K.); (L.L.); (M.R.A.)
- ICES, Toronto, ON M4N 3M5, Canada; (M.C.C.); (P.C.A.)
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Endocrinology, Women’s College Hospital, Toronto, ON M5S 1B2, Canada
| | - Mohammad R. Akbari
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON M5S 1B2, Canada; (V.G.); (J.K.); (L.L.); (M.R.A.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Peter C. Austin
- ICES, Toronto, ON M4N 3M5, Canada; (M.C.C.); (P.C.A.)
- Institute of Health Policy Management and Evaluation, University of Toronto Ontario Canada, Toronto, ON M5T 3M6, Canada
| | - Steven A. Narod
- Women’s College Research Institute, Women’s College Hospital, Toronto, ON M5S 1B2, Canada; (V.G.); (J.K.); (L.L.); (M.R.A.)
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON M5T 3M7, Canada; (J.D.B.); (L.R.)
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence: ; Tel.: +1-416-351-3765; Fax: +1-416-351-3767
| |
Collapse
|
59
|
Giannakeas V, Kotsopoulos J, Cheung MC, Rosella L, Brooks JD, Lipscombe L, Akbari MR, Austin PC, Narod SA. Analysis of Platelet Count and New Cancer Diagnosis Over a 10-Year Period. JAMA Netw Open 2022; 5:e2141633. [PMID: 35015064 PMCID: PMC8753503 DOI: 10.1001/jamanetworkopen.2021.41633] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
IMPORTANCE Individuals with cancer often have an elevated platelet count at the time of diagnosis. The extent to which an elevated platelet count is an indicator of cancer is unclear. OBJECTIVE To evaluate the association of an elevated platelet count with a cancer diagnosis. DESIGN, SETTING, AND PARTICIPANTS This nested case-control study included Ontario residents enrolled in the provincial health insurance plan who had 1 or more routine complete blood count (CBC) tests performed between January 1, 2007, and December 31, 2017, with follow-up through December 31, 2018. Case patients were individuals with a new cancer diagnosis during the observation period. Eligible control individuals were cancer free before the date of diagnosis for a case patient to whom they were matched. One case patient was matched to 3 controls based on sex, age, and health care use patterns. Data were analyzed from September 24, 2020, to July 13, 2021. EXPOSURES Case patients and controls were assigned to 1 of 5 exposure groups based on age- and sex-specific platelet count distributions in the control population: very low (≤10th percentile), low (>10th to 25th percentile), medium (>25th to <75th percentile), high (75th to <90th percentile), and very high (≥90th percentile). MAIN OUTCOMES AND MEASURES Odds ratios (ORs) were estimated for specific cancer sites for each category of platelet count at intervals up to 10 years after a blood test. RESULTS Of the 8 917 187 eligible Ontario residents with a routine CBC record available, 4 971 578 (55.8%) were women; the median age at the first CBC was 46.4 years (IQR, 32.5-59.5 years). Among individuals with a routine CBC record available, 495 341 (5.6%) received a diagnosis of first primary cancer during the 10-year observation period. The OR for a solid tumor diagnosis associated with a very high platelet count vs a medium platelet count in the 6-month period before the diagnosis was 2.32 (95% CI, 2.28-2.35). A very high platelet count was associated with colon (OR, 4.38; 95% CI, 4.22-4.54), lung (OR, 4.37; 95% CI, 4.22-4.53), ovarian (OR, 4.62; 95% CI, 4.19-5.09), and stomach (OR, 4.27; 95% CI, 3.91-4.66) cancers. Odds ratios attenuated with increasing time from CBC test to cancer diagnosis. CONCLUSIONS AND RELEVANCE In this nested case-control study, an elevated platelet count was associated with increased risk of cancer at several sites. Our findings suggest that an elevated platelet count could potentially serve as a marker for the presence of some cancer types.
Collapse
Affiliation(s)
- Vasily Giannakeas
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Joanne Kotsopoulos
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Matthew C. Cheung
- ICES, Toronto, Ontario, Canada
- Division of Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Laura Rosella
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jennifer D. Brooks
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Lorraine Lipscombe
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology, Women’s College Hospital, Toronto, Ontario, Canada
| | - Mohammad R. Akbari
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Peter C. Austin
- ICES, Toronto, Ontario, Canada
- Institute of Health Policy Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Steven A. Narod
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
60
|
Luo Z, Jiao B, Huang T, Zhao H, Zhang G. What is the Role of the Preoperative Blood-Based Inflammation Biomarkers in the Prognosis of Upper Tract Urothelial Carcinoma With Radical Nephroureterectomy? A Single-Centre Retrospective Study. Technol Cancer Res Treat 2022; 21:15330338221095667. [PMID: 35585719 PMCID: PMC9127859 DOI: 10.1177/15330338221095667] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objective: To assess the prognostic value of preoperative blood-based inflammation biomarkers, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and lymphocyte-monocyte ratio (LMR), on the survival outcomes of patients with upper tract urothelial carcinoma (UTUC) following radical nephroureterectomy (RNU). Methods: We retrospectively studied the data of 172 patients who were diagnosed with UTUC after RNU during 2008 to 2018. We determined the cut-off value by using X-tile software. The area under the curve (AUC) and concordance index (C-index) were utilized to compare the predictive accuracy between subgroups. We also performed decision curve analysis (DCA) to evaluate the clinical net benefit of prognostic models. The Kaplan-Meier method and Cox proportional hazards regression models were performed to evaluate the association between these inflammation biomarkers and survival outcomes. Results: The median follow-up period was 45.5 (range: 1-143; interquartile range, IQR: 27-77) months. Kaplan-Meier analyses showed that a high NLR or PLR significantly reduced overall survival (OS), cancer-specific survival (CSS), recurrence-free survival (RFS) and metastasis-free survival (MFS), and a low LMR markedly decreased RFS and MFS. The multivariate Cox proportional hazards model revealed that the NLR plus PLR was an independent predictor of worse survival (all P < .05). Additionally, the AUC and C-index of the new prognostic models were the largest for the 1- to 5-year OS, CSS, RFS, and MFS were the largest. Conclusion: Our study confirms that the combination of preoperative NLR and PLR could be an independent risk factor for UTUC patients who have undergone RNU. The addition of NLR and PLR may improve the accuracy of current prognostic models and help guide clinical strategies in the treatment of UTUC.
Collapse
Affiliation(s)
- Zhenkai Luo
- 569810Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binbin Jiao
- 569810Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Department of Urology, 36635China-Japan Friendship Hospital, Beijing, China
| | - Tao Huang
- Department of Urology, 36635China-Japan Friendship Hospital, Beijing, China.,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Hang Zhao
- Department of Urology, 36635China-Japan Friendship Hospital, Beijing, China.,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Guan Zhang
- 569810Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China.,Department of Urology, 36635China-Japan Friendship Hospital, Beijing, China.,Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| |
Collapse
|
61
|
Faria AVS, Yu B, Mommersteeg M, de Souza-Oliveira PF, Andrade SS, Spaander MCW, de Maat MPM, Peppelenbosch MP, Ferreira-Halder CV, Fuhler GM. Platelet-dependent signaling and Low Molecular Weight Protein Tyrosine Phosphatase expression promote aggressive phenotypic changes in gastrointestinal cancer cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166280. [PMID: 34610471 DOI: 10.1016/j.bbadis.2021.166280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Over the last decades, some members of the protein tyrosine phosphatase family have emerged as cancer promoters. Among them, the Low Molecular Weight Protein Tyrosine Phosphatase (LMWPTP) has been described to be associated with colorectal cancer liver metastasis and poor prostate cancer prognosis. Of importance in the process of cancer progression and metastasis is the interaction between tumor cells and platelets, as the latter are thought to promote several tumor hallmarks. Here, we examine to what extent LMWPTP expression in tumor cells affects their interaction with platelets. We demonstrate that the gene encoding LMWPTP is overexpressed in upper gastrointestinal (GI) cancer cell as well as colorectal cancer, and subsequently employ cell line models to show that the level of this phosphatase may be further augmented in the presence of platelets. We demonstrate that tumor-platelet interaction promotes GI tumor cell proliferation. Additionally, using know-down/-out models we show that LMWPTP expression in cancer cells contributes to a more efficient interaction with platelets and drives platelet-induced proliferation. These data are the first to demonstrate that phosphatases play a positive role in the tumor-promoting activities of platelets, with LMWPTP emerging as a key player promoting oncogenic phenotypic changes in tumor cells.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands; Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil
| | - Bingting Yu
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Michiel Mommersteeg
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | | | | | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Moniek P M de Maat
- Department of Hematology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil.
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
62
|
Tanaka H, Horioka K, Hasebe T, Sawada K, Nakajima S, Konishi H, Isozaki S, Goto M, Fujii Y, Kamikokura Y, Ogawa K, Nishikawa Y. Treatment of hepatocellular carcinoma with autologous platelets encapsulating sorafenib or lenvatinib: A novel therapy exploiting tumor‐platelet interactions. Int J Cancer 2021; 150:1640-1653. [DOI: 10.1002/ijc.33915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 12/25/2022]
Affiliation(s)
- Hiroki Tanaka
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Kie Horioka
- Department of Oncology‐Pathology Karolinska Institutet Stockholm Sweden
- Department of Legal Medicine International University of Health and Welfare Narita Japan
| | - Takumu Hasebe
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Koji Sawada
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Shunsuke Nakajima
- Department of Emergency Medicine Asahikawa Medical University Asahikawa Japan
| | - Hiroaki Konishi
- Department of Gastroenterology and Advanced Medical Sciences Asahikawa Medical University Asahikawa Japan
| | - Shotaro Isozaki
- Division of Metabolism and Biosystemic Science, Gastroenterology and Hematology/Oncology, Department of Medicine Asahikawa Medical University Asahikawa Japan
| | - Masanori Goto
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Yumiko Fujii
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | - Yuki Kamikokura
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| | | | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology Asahikawa Medical University Asahikawa Japan
| |
Collapse
|
63
|
Ielasi L, Tovoli F, Tonnini M, Tortora R, Magini G, Sacco R, Pressiani T, Trevisani F, Sansone V, Marasco G, Piscaglia F, Granito A. Beneficial Prognostic Effects of Aspirin in Patients Receiving Sorafenib for Hepatocellular Carcinoma: A Tale of Multiple Confounders. Cancers (Basel) 2021; 13:cancers13246376. [PMID: 34944996 PMCID: PMC8699252 DOI: 10.3390/cancers13246376] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/10/2021] [Accepted: 12/10/2021] [Indexed: 02/07/2023] Open
Abstract
Case-control observational studies suggested that aspirin might prevent hepatocellular carcinoma (HCC) in high-risk patients, even if randomized clinical trials are lacking. Information regarding aspirin in subjects who already developed HCC, especially in its advanced stage, are scarce. While aspirin might be a low-cost option to improve the prognosis, multiple confounders and safety concerns are to be considered. In our retrospective analyses of a prospective dataset (n = 699), after assessing the factors associated with aspirin prescription, we applied an inverse probability treatment weight analysis to address the prescription bias. Analyses of post-sorafenib survival were also performed to reduce the influence of subsequent medications. Among the study population, 133 (19%) patients were receiving aspirin at the time of sorafenib prescription. Aspirin users had a higher platelet count and a lower prevalence of esophageal varices, macrovascular invasion, and Child-Pugh B status. The benefit of aspirin was confirmed in terms of overall survival (HR 0.702, 95% CI 0.543-0.908), progression-free survival, disease control rate (58.6 vs. 49.5%, p < 0.001), and post-sorafenib survival even after weighting. Minor bleeding events were more frequent in the aspirin group. Aspirin use was associated with better outcomes, even after the correction for confounders. While safety concerns arguably remain a problem, prospective trials for patients at low risk of bleeding are warranted.
Collapse
Affiliation(s)
- Luca Ielasi
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
| | - Francesco Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (F.T.); (G.M.)
- Correspondence: ; Tel.: +39-051-214-2214; Fax: +39-051-214-2725
| | - Matteo Tonnini
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
| | - Raffaella Tortora
- Liver Unit, Department of Transplantation, Cardarelli Hospital, 80131 Naples, Italy;
| | - Giulia Magini
- Department of Gastroenterology and Transplant Hepatology, Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy;
| | - Rodolfo Sacco
- Gastroenterology Unit, Azienda Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy;
- Gastroenterology and Digestive Endoscopy Unit, Foggia University Hospital, 71122 Foggia, Italy
| | - Tiziana Pressiani
- Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy;
| | - Franco Trevisani
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (F.T.); (G.M.)
- Semeiotica Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Vito Sansone
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
| | - Giovanni Marasco
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (F.T.); (G.M.)
- Internal Medicine and Digestive Physiopathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (F.T.); (G.M.)
| | - Alessandro Granito
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (L.I.); (M.T.); (V.S.); (F.P.); (A.G.)
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (F.T.); (G.M.)
| |
Collapse
|
64
|
Wang Y, Xu X, Chen X, Li J. Multifunctional Biomedical Materials Derived from Biological Membranes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 34:e2107406. [PMID: 34739155 DOI: 10.1002/adma.202107406] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/24/2021] [Indexed: 02/06/2023]
Abstract
The delicate structure and fantastic functions of biological membranes are the successful evolutionary results of a long-term natural selection process. Their excellent biocompatibility and biofunctionality are widely utilized to construct multifunctional biomedical materials mainly by directly camouflaging materials with single or mixed biological membranes, decorating or incorporating materials with membrane-derived vesicles (e.g., exosomes), and designing multifunctional materials with the structure/functions of biological membranes. Here, the structure-function relationship of some important biological membranes and biomimetic membranes are discussed, such as various cell membranes, extracellular vesicles, and membranes from bacteria and organelles. Selected literature examples of multifunctional biomaterials derived from biological membranes for biomedical applications, such as drug- and gene-delivery systems, tissue-repair scaffolds, bioimaging, biosensors, and biological detection, are also highlighted. These designed materials show excellent properties, such as long circulation time, disease-targeted therapy, excellent biocompatibility, and selective recognition. Finally, perspectives and challenges associated with the clinical applications of biological-membrane-derived materials are discussed.
Collapse
Affiliation(s)
- Yuemin Wang
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xinyuan Xu
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
| | - Xingyu Chen
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- College of Medicine Southwest Jiaotong University Chengdu 610003 China
| | - Jianshu Li
- College of Polymer Science and Engineering State Key Laboratory of Polymer Materials Engineering Sichuan University Chengdu 610065 P. R. China
- State Key Laboratory of Oral Diseases West China Hospital of Stomatology Med‐X Center for Materials Sichuan University Chengdu 610041 China
| |
Collapse
|
65
|
Liu QL, Zhang Z, Wei X, Zhou ZG. Noncoding RNAs in tumor metastasis: molecular and clinical perspectives. Cell Mol Life Sci 2021; 78:6823-6850. [PMID: 34499209 PMCID: PMC11073083 DOI: 10.1007/s00018-021-03929-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/12/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023]
Abstract
Metastasis is the main culprit of cancer-associated mortality and involves a complex and multistage process termed the metastatic cascade, which requires tumor cells to detach from the primary site, intravasate, disseminate in the circulation, extravasate, adapt to the foreign microenvironment, and form organ-specific colonization. Each of these processes has been already studied extensively for molecular mechanisms focused mainly on protein-coding genes. Recently, increasing evidence is pointing towards RNAs without coding potential for proteins, referred to as non-coding RNAs, as regulators in shaping cellular activity. Since those first reports, the detection and characterization of non-coding RNA have explosively thrived and greatly enriched the understanding of the molecular regulatory networks in metastasis. Moreover, a comprehensive description of ncRNA dysregulation will provide new insights into novel tools for the early detection and treatment of metastatic cancer. In this review, we focus on discussion of the emerging role of ncRNAs in governing cancer metastasis and describe step by step how ncRNAs impinge on cancer metastasis. In particular, we highlight the diagnostic and therapeutic applications of ncRNAs in metastatic cancer.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhe Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Zong-Guang Zhou
- Department of Gastrointestinal Surgery, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
66
|
Dombroski JA, Hope JM, Sarna NS, King MR. Channeling the Force: Piezo1 Mechanotransduction in Cancer Metastasis. Cells 2021; 10:2815. [PMID: 34831037 PMCID: PMC8616475 DOI: 10.3390/cells10112815] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/08/2021] [Accepted: 10/12/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer metastasis is one of the leading causes of death worldwide, motivating research into identifying new methods of preventing cancer metastasis. Recently there has been increasing interest in understanding how cancer cells transduce mechanical forces into biochemical signals, as metastasis is a process that consists of a wide range of physical forces. For instance, the circulatory system through which disseminating cancer cells must transit is an environment characterized by variable fluid shear stress due to blood flow. Cancer cells and other cells can transduce physical stimuli into biochemical responses using the mechanosensitive ion channel Piezo1, which is activated by membrane deformations that occur when cells are exposed to physical forces. When active, Piezo1 opens, allowing for calcium flux into the cell. Calcium, as a ubiquitous second-messenger cation, is associated with many signaling pathways involved in cancer metastasis, such as angiogenesis, cell migration, intravasation, and proliferation. In this review, we discuss the roles of Piezo1 in each stage of cancer metastasis in addition to its roles in immune cell activation and cancer cell death.
Collapse
Affiliation(s)
| | | | | | - Michael R. King
- King Lab, Department of Biomedical Engineering, Vanderbilt University, 5824 Stevenson Center, Nashville, TN 37235, USA; (J.A.D.); (J.M.H.); (N.S.S.)
| |
Collapse
|
67
|
Liu Y, Zhang Y, Ding Y, Zhuang R. Platelet-mediated tumor metastasis mechanism and the role of cell adhesion molecules. Crit Rev Oncol Hematol 2021; 167:103502. [PMID: 34662726 DOI: 10.1016/j.critrevonc.2021.103502] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/17/2021] [Accepted: 10/10/2021] [Indexed: 12/12/2022] Open
Abstract
Mounting evidence suggests that platelets play an essential role in cancer metastasis. The interactions between platelets and circulating tumor cells (CTCs) promote cancer metastasis. CTCs induce platelet activation and aggregation, and activated platelets gather and protect CTCs from shear stress and natural killer cells. Finally, platelets stimulate CTC anoikis resistance, epithelial-to-mesenchymal transition, angiogenesis, extravasation, and eventually, metastasis. Cell adhesion molecules (CAMs) have been identified as active players during the interaction of CTCs with platelets, but the specific mechanism underlying the contribution of platelet-associated CAMs to CTC metastasis remains unclear. In this review, we introduce the mechanism of platelet-related tumor metastasis and particularly focus on the role of CAMs in it.
Collapse
Affiliation(s)
- Yitian Liu
- Department of Immunology, the Fourth Military Medical University, #169 Changlexilu Road, Xi'an, Shaanxi, 710032, China; Orthopedic Department of Tangdu Hospital, the Fourth Military Medical University, #1 Xinsi Road, Xi'an, Shaanxi, 710032, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, #127 Youyixilu Road, Xi'an, Shaanxi, 710072, China
| | - Yong Ding
- Orthopedic Department of Tangdu Hospital, the Fourth Military Medical University, #1 Xinsi Road, Xi'an, Shaanxi, 710032, China
| | - Ran Zhuang
- Department of Immunology, the Fourth Military Medical University, #169 Changlexilu Road, Xi'an, Shaanxi, 710032, China; Institute of Medical Research, Northwestern Polytechnical University, #127 Youyixilu Road, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
68
|
Kubota K, Shimizu A, Notake T, Masuo H, Hosoda K, Yasukawa K, Hayashi H, Umemura K, Kamachi A, Goto T, Tomida H, Yamazaki S, Soejima Y. Preoperative Peripheral Blood Lymphocyte-to-Monocyte Ratio Predicts Long-Term Outcome for Patients with Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2021; 29:1437-1448. [PMID: 34664139 DOI: 10.1245/s10434-021-10848-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although various biomarkers are useful in predicting cancer prognosis, the most effective preoperative systemic biomarkers for pancreatic ductal adenocarcinoma (PDAC) have not been established. This study aimed to evaluate whether the lymphocyte-to-monocyte ratio (LMR) can predict the long-term outcomes for patients who were to undergo surgical resection of PDAC. METHODS The study involved 170 patients with PDAC who underwent resection. Multivariate analysis was performed to identify the independent prognostic factors for overall survival (OS) and disease-free survival (DFS) among clinicopathologic, surgical, and seven systemic biomarker-related factors including LMR. Subgroup analysis of PDAC located in the body and tail of the pancreas (B/T PDAC) was performed (n = 60) to eliminate the influence of preoperative cholangitis and surgical procedure. Furthermore, OS according to the postoperative course of the LMR value group was investigated. RESULTS A low LMR (<3.3) was the only independent predictive factor for OS (hazard ratio [HR], 3.52; p < 0.001) and DFS (HR, 3.31; p < 0.001) among the systemic biomarkers. Subgroup analysis of the B/T PDAC also showed that low the LMR was the independent predictive factor for OS (HR, 3.24; p = 0.002) and DFS (HR, 4.42; p = 0.003). The PDAC that maintained a high LMR from before surgery to 1 year after surgery showed good long-term outcomes (median OS, 8.5 years; 5-year survival rate, 61.8 %). CONCLUSIONS Preoperative LMR was an independent predictor of OS and DFS after surgery for PDAC. Maintaining a high LMR through the pre- and postoperative courses might improve the prognosis for patients with PDAC.
Collapse
Affiliation(s)
- Koji Kubota
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Akira Shimizu
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan.
| | - Tsuyoshi Notake
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hitoshi Masuo
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Kiyotaka Hosoda
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Koya Yasukawa
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hikaru Hayashi
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Kentaro Umemura
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Atsushi Kamachi
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takamune Goto
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hidenori Tomida
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Shiori Yamazaki
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yuji Soejima
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
69
|
Ma Y, Li G, Yu M, Sun X, Nian J, Gao Y, Li X, Ding T, Wang X. Prognostic significance of thrombocytosis in lung cancer: a systematic review and meta-analysis. Platelets 2021; 32:919-927. [PMID: 32892682 DOI: 10.1080/09537104.2020.1810653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A potential relationship between poor prognosis and thrombocytosis has been suggested by previous studies in lung cancer, but the conclusions continued to be controversial. Here, we performed a meta-analysis to explore the prognostic impact of thrombocytosis in lung cancer. The Cochrane Library, EMBASE and PubMed databases were comprehensively and systematically retrieved from establishment to May 5, 2020. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) were applied to evaluate overall effects. Heterogeneity was assessed using I2 statistics and Cochran's Q test. Sensitivity and subgroup analyses were performed to analyze the sources of heterogeneity. Publication bias was examined using the Egger's test and pooled HR was regulated using the trim-and-fill approach when publication bias was observed. A total of 37 studies including 14,833 patients were enrolled in the meta-analysis. Thrombocytosis was significantly correlated to poor overall survival (HR 1.033; 95% CI 1.017-1.050), disease-free survival (HR 1.568; 95% CI 1.276-1.928), and progression-free survival (HR 1.653; 95% CI 1.069-2.556). Although publication bias was identified, rectification for this bias using the trim-and-fill approach did not change the combined HR substantially. In conclusion, this meta-analysis result suggested that thrombocytosis is a predictor of poor prognosis in lung cancer.
Collapse
Affiliation(s)
- Yunfei Ma
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Guangda Li
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
| | - Mingwei Yu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xu Sun
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Jiayun Nian
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yu Gao
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoxiao Li
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Tongjing Ding
- School of Graduates, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaomin Wang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
70
|
Abstract
Platelets evolved from nucleated thrombocytes that exhibit both coagulation and immune function. The essential role of platelets in coagulation is common knowledge. The larger and critical role of platelets in immune responses and cancer are frequently overlooked in our modern-day, large-data-set, sequencing-oriented efforts. Much like Waldo, their small size, biophysical characteristics, rapid biological responses, active cytoskeleton, migration capacity, and lack of a nucleus make them difficult to track as single platelets disappear while executing their function into the histologic "tissue scape". The adaptive evolution of platelets is linked to placentalization and stopping massive blood loss. This resulted in exclusion of any platelet nucleus and therefore sustainable gene expression due to being extruded in the billions (1011) per day from megakaryocytes under bone marrow protection. The platelets' small size and sheer number in circulation, combined with an active open canalicular exchange- and membrane-reserve system, plus an array of pathogen receptors enable them to deal with small pathogenic viral treats and to decorate larger ones for further immune identification and immune-cell recruitment. Once stimulated, platelets release most serum-based cytokines and growth factors that contribute to cell growth and wound repair, and potentially to immune suppression. From a self-taught practitioner of the illustrative arts with a ken for platelet biology, this offering is a humble attempt to provide a stimulating sketch of the critical importance of platelet biology and insights into potential new directions for finding the Waldo-esque platelet.
Collapse
Affiliation(s)
- D G Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
71
|
Wassmer SC, Humpel C, Orian JM. Editorial: Platelets as Players in Neuropathologies: Novel Diagnostic and Therapeutic Targets. Front Immunol 2021; 12:772352. [PMID: 34659273 PMCID: PMC8513524 DOI: 10.3389/fimmu.2021.772352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Samuel C. Wassmer
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christian Humpel
- Laboratory for Psychiatry and Experimental Alzheimer’s Research, Medical University Innsbruck, Innsbruck, Austria
| | - Jacqueline M. Orian
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| |
Collapse
|
72
|
Circulating Tumour Cell Numbers Correlate with Platelet Count and Circulating Lymphocyte Subsets in Men with Advanced Prostate Cancer: Data from the ExPeCT Clinical Trial (CTRIAL-IE 15-21). Cancers (Basel) 2021; 13:cancers13184690. [PMID: 34572916 PMCID: PMC8466183 DOI: 10.3390/cancers13184690] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Cancer cells (CTCs) can be found in the bloodstream in men with advanced prostate cancer. Blood platelets, which normally help the blood to clot, may help the cancer cells to spread throughout the body by preventing the body’s immune system from finding and destroying them while they are in the bloodstream. Blood samples were taken from men with prostate cancer who were involved in the ExPeCT clinical trial, some of whom were taking part in a regular exercise programme. The numbers of CTCs, platelets and immune system cells were counted and compared. Blood samples with more CTCs had higher numbers of platelets and higher numbers of some types of immune system cells. Some differences were also found in men involved in the exercise programme. This study helps to show that CTCs numbers are related to platelet and immune cell numbers in the blood. Abstract Interactions between circulating tumour cells (CTCs) and platelets are thought to inhibit natural killer(NK)-cell-induced lysis. We attempted to correlate CTC numbers in men with advanced prostate cancer with platelet counts and circulating lymphocyte numbers. Sixty-one ExPeCT trial participants, divided into overweight/obese and normal weight groups on the basis of a BMI ≥ 25 or <25, were randomized to participate or not in a six-month exercise programme. Blood samples at randomization, and at three and six months, were subjected to ScreenCell filtration, circulating platelet counts were obtained, and flow cytometry was performed on a subset of samples (n = 29). CTC count positively correlated with absolute total lymphocyte count (r2 = 0.1709, p = 0.0258) and NK-cell count (r2 = 0.49, p < 0.0001). There was also a positive correlation between platelet count and CTC count (r2 = 0.094, p = 0.0001). Correlation was also demonstrated within the overweight/obese group (n = 123, p < 0.0001), the non-exercise group (n = 79, p = 0.001) and blood draw samples lacking platelet cloaking (n = 128, p < 0.0001). By flow cytometry, blood samples from the exercise group (n = 15) had a higher proportion of CD3+ T-lymphocytes (p = 0.0003) and lower proportions of B-lymphocytes (p = 0.0264) and NK-cells (p = 0.015) than the non-exercise group (n = 14). These findings suggest that CTCs engage in complex interactions with the coagulation cascade and innate immune system during intravascular transit, and they present an attractive target for directed therapy at a vulnerable stage in metastasis.
Collapse
|
73
|
Zhang J, Li F, Augi T, Williamson KM, Onishi K, Hogan MV, Neal MD, Wang JHC. Platelet HMGB1 in Platelet-Rich Plasma (PRP) promotes tendon wound healing. PLoS One 2021; 16:e0251166. [PMID: 34529657 PMCID: PMC8445483 DOI: 10.1371/journal.pone.0251166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
Platelet-rich plasma (PRP) is a widely used autologous treatment for tendon injuries in clinics. Platelets (PLTs) are a major source of high mobility group box1 (HMGB1) that is gaining attention as a chemoattractant that can recruit stem cells to the wound area to enhance healing of injured tissues; however, the contribution of PLT HMGB1 in wounded tendon healing remains unexplored. This study investigated the effect of PLT HMGB1 within PRP on tendon healing using PLT HMGB1 knockout (KO) and GFP mice. A window defect was created in the patellar tendons of both groups of mice, and wounds were treated with either saline, PRP isolated from PLT HMGB1-KO mice, or PRP isolated from GFP mice. Seven days post-treatment, animals were sacrificed and analyzed by gross inspection, histology, and immunostaining for characteristic signs of tendon healing and repair. Our results showed that in comparison to mice treated with PRP from PLT HMGB1-KO mice, wounds treated with PRP from GFP mice healed faster and exhibited a better organization in tendon structure. Mice treated with PRP from PLT HMGB1-KO mice produced tendon tissue with large premature wound areas and low cell densities. However, wounds of PLT HMGB1-KO mice showed better healing with PRP from HMGB1-KO mice compared to saline treatment. Moreover, wounds treated with PRP from GFP mice had increased extracellular HMGB1, decreased CD68, increased stem cell markers CD146 and CD73, and increased collagen III protein expression levels compared to those treated with PRP from PLT HMGB1-KO mice. Thus, PLT HMGB1 within PRP plays an important role in tendon wound healing by decreasing inflammation, increasing local HMGB1 levels, and recruiting stem cells to the wound area in the tendon. Our findings also suggest that the efficacy of PRP treatment for tendon injuries in clinics may depend on PLT HMGB1 within PRP preparations.
Collapse
Affiliation(s)
- Jianying Zhang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Feng Li
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Tyler Augi
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kelly M. Williamson
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kentaro Onishi
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - MaCalus V. Hogan
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - James H.-C. Wang
- MechanoBiology Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
74
|
Schneider MA, Heeb L, Beffinger MM, Pantelyushin S, Linecker M, Roth L, Lehmann K, Ungethüm U, Kobold S, Graf R, van den Broek M, Vom Berg J, Gupta A, Clavien PA. Attenuation of peripheral serotonin inhibits tumor growth and enhances immune checkpoint blockade therapy in murine tumor models. Sci Transl Med 2021; 13:eabc8188. [PMID: 34524861 DOI: 10.1126/scitranslmed.abc8188] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marcel André Schneider
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Laura Heeb
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Michal Mateusz Beffinger
- Institute of Laboratory Animal Science, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Stanislav Pantelyushin
- Institute of Laboratory Animal Science, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Michael Linecker
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Lilian Roth
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland.,Surgical Oncology Research Laboratory, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Kuno Lehmann
- Surgical Oncology Research Laboratory, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Udo Ungethüm
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Ludwig-Maximilians-Universität München, Lindwurmstrasse 2a, D-80337 Munich, Germany.,German Center for Translational Cancer Research (DKTK), partner site Munich, Pettenkoferstr. 8a, D-80336 Munich, Germany
| | - Rolf Graf
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zürich, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Johannes Vom Berg
- Institute of Laboratory Animal Science, University of Zürich, Wagistrasse 12, CH-8952 Schlieren, Switzerland
| | - Anurag Gupta
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| | - Pierre-Alain Clavien
- Laboratory of the Swiss Hepato-Pancreatico-Biliary (HPB) and Transplantation Centre, Department of Surgery, University Hospital and University of Zürich, Raemistrasse 100, CH-8091 Zürich, Switzerland
| |
Collapse
|
75
|
Caruntu A, Moraru L, Lupu M, Taubner L, Caruntu C, Tanase C. The Hidden Treasures of Preoperative Blood Assessment in Oral Cancer: A Potential Source of Biomarkers. Cancers (Basel) 2021; 13:cancers13174475. [PMID: 34503285 PMCID: PMC8430940 DOI: 10.3390/cancers13174475] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary In this study, we assess the prognostic potential of immune and inflammatory elements determined preoperatively in the peripheral blood of patients with oral squamous cell carcinoma (OSCC). Preoperative plasma fibrinogen (Fib) and platelet-to-lymphocyte ratio (PLR) show strong correlations with patients’ outcomes. Analyzed together, in a new parameter named Fibrinogen-PLR Algorithm (FiPLA), predictive power increases significantly. Clinicians can use this new, easy, cost-effective, and globally available tool for risk stratification of patients with OSCC, as early as from the moment of diagnosis. Abstract (1) Background: Oral squamous cell carcinoma (OSCC) is a common malignancy, and the impact of immune and inflammatory mechanisms in its development and progression are of major interest. The aim of our study is to assess the prognostic potential of circulating immune and inflammatory elements determined preoperatively in patients with OSCC, as well as the development of a new compound parameter with predictive value. (2) Methods: We assessed preoperative fibrinogen (Fib) and the platelet-to-lymphocyte ratio (PLR) in 111 OSCC patients. Using a mathematic algorithm, we determined a composite parameter with cumulative information from Fib and PLR, named Fibrinogen-PLR Algorithm (FiPLA). Survival analysis, followed by bivariate and multivariate analyses, was subsequently conducted. (3) Results: Increased preoperative Fib and PLR levels were associated with poor outcome in OSCC (p = 0.0001 and p = 0.0015, respectively). Preoperative FiPLA values were also associated with poor patient survival (p < 0.0001). Multivariate analysis confirmed the independent prognostic role for FiPLA only (CI95% 1.232–67.770, p = 0.03), showing the superior predictive value of FiPLA compared to its individual components. (4) Conclusions: Preoperative assessments of circulating immune and inflammatory elements can provide high-quality prognostic information, and they represent valuable tools in clinical practice, facilitating the early risk stratification of patients with OSCC.
Collapse
Affiliation(s)
- Ana Caruntu
- Department of Oral and Maxillofacial Surgery, Carol Davila Central Military Emergency Hospital, 010825 Bucharest, Romania; (A.C.); (L.M.)
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Liliana Moraru
- Department of Oral and Maxillofacial Surgery, Carol Davila Central Military Emergency Hospital, 010825 Bucharest, Romania; (A.C.); (L.M.)
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Mihai Lupu
- Dermatology Research Laboratory, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Lacramioara Taubner
- Clinical Laboratory, Carol Davila Central Military Emergency Hospital, 010825 Bucharest, Romania;
| | - Constantin Caruntu
- Department of Physiology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Dermatology, Prof. N.C. Paulescu National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
- Correspondence: ; Tel.: +40-745-086-978
| | - Cristiana Tanase
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, 050096 Bucharest, Romania;
- Proteomics Department, Cajal Institute, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| |
Collapse
|
76
|
Rashdan S, Iyengar P, Minna JD, Gerber DE. Narrative review: molecular and genetic profiling of oligometastatic non-small cell lung cancer. Transl Lung Cancer Res 2021; 10:3351-3368. [PMID: 34430372 PMCID: PMC8350108 DOI: 10.21037/tlcr-21-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/28/2021] [Indexed: 11/30/2022]
Abstract
Objective The objectives of this review are to discuss: the definition, clinical and biologic features of oligometastatic non-small cell lung cancer (NSCLC), as well as the concept of treating oligoprogression in oligometastatic NSCLC. Background A substantial proportion of patients diagnosed with lung cancer present with metastatic disease, and a large portion of patients who present with localized disease later develop metastases. Oligometastatic NSCLC is defined as an intermediate state between localized and widespread metastatic disease, where there may be a role for curative localized therapy approach by treating the primary tumor and all metastases with radiotherapy or surgery. Despite the increasing application of this approach in patients with lung cancer, the identification of patients who might benefit from this approach is yet to be well characterized. Methods After a systematic review of the literature, a PubMed search was performed using the English language and the key terms: oligometastatic, non-small cell lung cancer (NSCLC), localized consolidative treatment (LCT), biomarkers, biologic features, clinical features. Over 500 articles were retrieved between 1889–2021. A total of 178 papers discussing the definition, clinical and biologic factors leading to oligometastatic NSCLC were reviewed and included in the discussion of this paper. Conclusions Oligometastatic NSCLC is a unique entity. Identifying patients who have oligometastatic NSCLC accurately using a combination of clinical and biologic features and treating them with localized consolidative approach appropriately results in improvement of outcome. Further understanding of the molecular mechanisms driving the formation of oligometastatic NSCLC is an important area of focus for future studies.
Collapse
Affiliation(s)
- Sawsan Rashdan
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology-Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Puneeth Iyengar
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John D Minna
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology-Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - David E Gerber
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology-Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Population and Data Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
77
|
Abstract
IMPORTANCE Individuals with newly diagnosed cancer often have a high platelet count (thrombocytosis). Whether thrombocytosis is associated with the presence of an undiagnosed cancer remains unknown. OBJECTIVE To assess whether a new diagnosis of thrombocytosis is associated with a subsequent risk of cancer among adults. DESIGN, SETTING, AND PARTICIPANTS This population-based retrospective cohort study was conducted using linked laboratory data from Ontario, Canada, from January 1, 2007, to December 31, 2017, with follow-up until December 31, 2018. The study cohort included adults aged 40 to 75 years on the date of a routine complete blood count (CBC) test (index test) who had a normal platelet count in the 2 previous years and no history of cancer. Data analysis was performed in December 2020. EXPOSURES Exposed individuals were those with a platelet count greater than 450 × 109/L. Matched unexposed control individuals had a platelet count within the reference range (150 × 109/L to 450 × 109/L) reported within 30 days of the exposure. MAIN OUTCOMES AND MEASURES Incident cancers within 5 years after diagnosis of thrombocytosis. Absolute and relative risks for cancer associated with thrombocytosis were estimated for all cancers and for cancers at specific sites. RESULTS Of the 3 386 716 Ontario residents with a recorded routine CBC test result, 53 339 (1.6%) had thrombocytosis and a prior normal platelet count. Among individuals with thrombocytosis, the median age was 59.7 years (interquartile range, 50.2-67.4 years) and 37 349 (70.0%) were women. Among the 51 624 individuals with thrombocytosis included in the matched analysis, 2844 (5.5%) had received a diagnosis of a solid cancer in the 2-year follow-up period and 3869 (7.5%) had received a diagnosis within 5 years. The relative risk (RR) for developing any solid cancer within 2 years was 2.67 (95% CI, 2.56-2.79). Associations were found between thrombocytosis and cancers of the ovary (RR, 7.11; 95% CI, 5.59-9.03), stomach (RR, 5.53; 95% CI, 4.12-7.41), colon (RR, 5.41; 95% CI, 4.80-6.10), lung (RR, 4.41; 95% CI, 4.02-4.85), kidney (RR, 3.64; 95% CI, 2.94-4.51), and esophagus (RR, 3.64; 95% CI, 2.46-5.40). CONCLUSIONS AND RELEVANCE In this cohort study, an increased platelet count was associated with an increased risk of cancer for at least 2 years. The results suggest that individuals with unexplained thrombocytosis should be offered screening for several cancers.
Collapse
Affiliation(s)
- Vasily Giannakeas
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- ICES, Toronto, Ontario, Canada
| | - Steven A Narod
- Women's College Research Institute, Women's College Hospital, Toronto, Ontario, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
78
|
Kanikarla Marie P, Fowlkes NW, Afshar-Kharghan V, Martch SL, Sorokin A, Shen JP, Morris VK, Dasari A, You N, Sood AK, Overman MJ, Kopetz S, Menter DG. The Provocative Roles of Platelets in Liver Disease and Cancer. Front Oncol 2021; 11:643815. [PMID: 34367949 PMCID: PMC8335590 DOI: 10.3389/fonc.2021.643815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Both platelets and the liver play important roles in the processes of coagulation and innate immunity. Platelet responses at the site of an injury are rapid; their immediate activation and structural changes minimize the loss of blood. The majority of coagulation proteins are produced by the liver—a multifunctional organ that also plays a critical role in many processes: removal of toxins and metabolism of fats, proteins, carbohydrates, and drugs. Chronic inflammation, trauma, or other causes of irreversible damage to the liver can dysregulate these pathways leading to organ and systemic abnormalities. In some cases, platelet-to-lymphocyte ratios can also be a predictor of disease outcome. An example is cirrhosis, which increases the risk of bleeding and prothrombotic events followed by activation of platelets. Along with a triggered coagulation cascade, the platelets increase the risk of pro-thrombotic events and contribute to cancer progression and metastasis. This progression and the resulting tissue destruction is physiologically comparable to a persistent, chronic wound. Various cancers, including colorectal cancer, have been associated with increased thrombocytosis, platelet activation, platelet-storage granule release, and thrombosis; anti-platelet agents can reduce cancer risk and progression. However, in cancer patients with pre-existing liver disease who are undergoing chemotherapy, the risk of thrombotic events becomes challenging to manage due to their inherent risk for bleeding. Chemotherapy, also known to induce damage to the liver, further increases the frequency of thrombotic events. Depending on individual patient risks, these factors acting together can disrupt the fragile balance between pro- and anti-coagulant processes, heightening liver thrombogenesis, and possibly providing a niche for circulating tumor cells to adhere to—thus promoting both liver metastasis and cancer-cell survival following treatment (that is, with minimal residual disease in the liver).
Collapse
Affiliation(s)
- Preeti Kanikarla Marie
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalie W Fowlkes
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephanie L Martch
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexey Sorokin
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Van K Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nancy You
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David George Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
79
|
CD8 + T cells inhibit metastasis and CXCL4 regulates its function. Br J Cancer 2021; 125:176-189. [PMID: 33795809 PMCID: PMC8292398 DOI: 10.1038/s41416-021-01338-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/21/2021] [Accepted: 02/23/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND The mechanism by which immune cells regulate metastasis is unclear. Understanding the role of immune cells in metastasis will guide the development of treatments improving patient survival. METHODS We used syngeneic orthotopic mouse tumour models (wild-type, NOD/scid and Nude), employed knockout (CD8 and CD4) models and administered CXCL4. Tumours and lungs were analysed for cancer cells by bioluminescence, and circulating tumour cells were isolated from blood. Immunohistochemistry on the mouse tumours was performed to confirm cell type, and on a tissue microarray with 180 TNBCs for human relevance. TCGA data from over 10,000 patients were analysed as well. RESULTS We reveal that intratumoral immune infiltration differs between metastatic and non-metastatic tumours. The non-metastatic tumours harbour high levels of CD8+ T cells and low levels of platelets, which is reverse in metastatic tumours. During tumour progression, platelets and CXCL4 induce differentiation of monocytes into myeloid-derived suppressor cells (MDSCs), which inhibit CD8+ T-cell function. TCGA pan-cancer data confirmed that CD8lowPlatelethigh patients have a significantly lower survival probability compared to CD8highPlateletlow. CONCLUSIONS CD8+ T cells inhibit metastasis. When the balance between CD8+ T cells and platelets is disrupted, platelets produce CXCL4, which induces MDSCs thereby inhibiting the CD8+ T-cell function.
Collapse
|
80
|
Saha B, Mathur T, Tronolone JJ, Chokshi M, Lokhande GK, Selahi A, Gaharwar AK, Afshar-Kharghan V, Sood AK, Bao G, Jain A. Human tumor microenvironment chip evaluates the consequences of platelet extravasation and combinatorial antitumor-antiplatelet therapy in ovarian cancer. SCIENCE ADVANCES 2021; 7:eabg5283. [PMID: 34290095 PMCID: PMC8294767 DOI: 10.1126/sciadv.abg5283] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/04/2021] [Indexed: 05/13/2023]
Abstract
Platelets extravasate from the circulation into tumor microenvironment, enable metastasis, and confer resistance to chemotherapy in several cancers. Therefore, arresting tumor-platelet cross-talk with effective and atoxic antiplatelet agents in combination with anticancer drugs may serve as an effective cancer treatment strategy. To test this concept, we create an ovarian tumor microenvironment chip (OTME-Chip) that consists of a platelet-perfused tumor microenvironment and which recapitulates platelet extravasation and its consequences. By including gene-edited tumors and RNA sequencing, this organ-on-chip revealed that platelets and tumors interact through glycoprotein VI (GPVI) and tumor galectin-3 under shear. Last, as proof of principle of a clinical trial, we showed that a GPVI inhibitor, Revacept, impairs metastatic potential and improves chemotherapy. Since GPVI is an antithrombotic target that does not impair hemostasis, it represents a safe cancer therapeutic. We propose that OTME-Chip could be deployed to study other vascular and hematological targets in cancer.
Collapse
Affiliation(s)
- Biswajit Saha
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Tanmay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
| | - James J Tronolone
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Mithil Chokshi
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Giriraj K Lokhande
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Amirali Selahi
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
- Materials Science and Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA
- Center for Remote Health Technologies and Systems, Texas A&M University, College Station TX 77840, USA
| | - Vahid Afshar-Kharghan
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gang Bao
- Department of Bioengineering, George R. Brown School of Engineering, Rice University, Houston, TX 77005, USA
| | - Abhishek Jain
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX 77840, USA.
- Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
- Department of Cardiovascular Sciences, Houston Methodist Academic Institute, Houston, TX 77030, USA
| |
Collapse
|
81
|
Camilli M, Iannaccone G, La Vecchia G, Cappannoli L, Scacciavillani R, Minotti G, Massetti M, Crea F, Aspromonte N. Platelets: the point of interconnection among cancer, inflammation and cardiovascular diseases. Expert Rev Hematol 2021; 14:537-546. [PMID: 34126832 DOI: 10.1080/17474086.2021.1943353] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The association between thrombosis, cancer and inflammation is well-established. Platelets play a major role in atherosclerosis, inflammation and immune response. Furthermore, growing evidence suggests that they are also significantly involved in tumor development and progression so that anti-platelet agents may prevent cancer and improve outcomes in oncological patients. In this review, we aimed at analyzing the relationship between platelets, cardiovascular diseases and cancer. A comprehensive study in the main educational platforms was performed and high-quality original articles and reviews were included. AREAS COVERED This review will focus on the role of platelets in cardiovascular disease and in cancer genesis and progression, analyzing their function as immune cells that link inflammation to thrombosis. Finally, it will examine the recent controversies on the use of anti-platelet agents as cancer medications, in particular the already known anti-tumor properties of aspirin, as well as the new perspectives regarding P2Y12 inhibitors. EXPERT OPINION Platelet-cancer crosstalk generates a vicious feed-back loop involving tumor cells and secreting molecules that activate platelets, which in turn promote cancer-associated inflammation, proliferation, spreading and immune system evasion. Therefore, platelets inhibition may represent an innovative therapeutical strategy offered to cancer patients, in the perspective of personalized medicine.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy
| | - Giulia Iannaccone
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy
| | - Giulia La Vecchia
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy
| | - Luigi Cappannoli
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy
| | - Roberto Scacciavillani
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy
| | - Giorgio Minotti
- Department of Medicine, Center for Integrated Research and Unit of Drug Sciences, University Campus Bio-Medico, Rome, Italy
| | - Massimo Massetti
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Nadia Aspromonte
- Department of Cardiovascular Sciences and Thoracic Sciences, Catholic University of the Sacred Heart, Roma, Italy.,Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
82
|
Platelet-to-lymphocyte ratio predicts therapy outcomes of transarterial chemoembolization plus apatinib in the treatment of advanced hepatocellular carcinoma. Anticancer Drugs 2021; 31:966-972. [PMID: 31977568 DOI: 10.1097/cad.0000000000000913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To evaluate the predictive value of preoperative biochemical marker [platelet-to-lymphocyte ratio (PLR)] in patients with advanced hepatocellular cancer receiving transarterial chemoembolization (TACE) plus targeted molecular therapy (apatinib) treatment. Clinical records of 134 patients receiving the treatment of TACE + apatinib (TACE-A) and the treatment of TACE alone were compared in a single-center study. Time to progression (TTP) and overall survival (OS) were compared between TACE-A and TACE alone groups in patients with PLR > 150 and PLR ≤ 150, respectively. The area under the receiver operating characteristic (ROC) curve was used to determine the prediction power of PLR. The median TTP and OS in the TACE-A group were significantly longer than those in the TACE alone group (P < 0.001). The median TTP and OS in the TACE-A (PLR ≤ 150) group were longer than those in the TACE-A (PLR > 150) group (P < 0.05). There was no significant difference between TACE-A (PLR > 150) and TACE alone (P = 0.232) groups in OS, but the median TTP in the TACE-A (PLR > 150) group was longer than that in the TACE alone group (P = 0.001). ROC analysis showed that the area under the curve was 0.643 and 0.623 for 6- and 12-month survival, respectively. PLR might predict the results of patients with advanced hepatocellular carcinoma received TACE-A treatment.
Collapse
|
83
|
Hamza MS, Mousa SA. Cancer-Associated Thrombosis: Risk Factors, Molecular Mechanisms, Future Management. Clin Appl Thromb Hemost 2021; 26:1076029620954282. [PMID: 32877229 PMCID: PMC7476343 DOI: 10.1177/1076029620954282] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Venous thromboembolism (VTE) is a major health problem in patients with cancer. Cancer augments thrombosis and causes cancer-associated thrombosis (CAT) and vice versa thrombosis amplifies cancer progression, termed thrombosis-associated cancer (TAC). Risk factors that lead to CAT and TAC include cancer type, chemotherapy, radiotherapy, hormonal therapy, anti-angiogenesis therapy, surgery, or supportive therapy with hematopoietic growth factors. There are some other factors that have an effect on CAT and TAC such as tissue factor, neutrophil extracellular traps (NETs) released in response to cancer, cancer procoagulant, and cytokines. Oncogenes, estrogen hormone, and thyroid hormone with its integrin αvβ3 receptor promote angiogenesis. Lastly, patient-related factors can play a role in development of thrombosis in cancer. Low-molecular-weight heparin and direct oral anticoagulants (DOACs) are used in VTE prophylaxis and treatment rather than vitamin K antagonist. Now, there are new directions for potential management of VTE in patients with cancer such as euthyroid, blockade of thyroid hormone receptor on integrin αvβ3, sulfated non-anticoagulant heparin, inhibition of NETs and stratifying low and high-risk patients with significant bleeding problems with DOACs.
Collapse
Affiliation(s)
- Marwa S. Hamza
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| | - Shaker A. Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
- Shaker A. Mousa, PhD, The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, 1 Discovery Drive, Rensselaer, NY 12144, USA.
| |
Collapse
|
84
|
Dymicka-Piekarska V, Koper-Lenkiewicz OM, Zińczuk J, Kratz E, Kamińska J. Inflammatory cell-associated tumors. Not only macrophages (TAMs), fibroblasts (TAFs) and neutrophils (TANs) can infiltrate the tumor microenvironment. The unique role of tumor associated platelets (TAPs). Cancer Immunol Immunother 2021; 70:1497-1510. [PMID: 33146401 PMCID: PMC8139882 DOI: 10.1007/s00262-020-02758-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
It is well known that various inflammatory cells infiltrate cancer cells. Next to TAMs (tumor-associated macrophages), TAFs (tumor-associated fibroblasts) and TANs (tumor-associated neutrophils) also platelets form the tumor microenvironment. Taking into account the role of platelets in the development of cancer, we have decided to introduce a new term: tumor associated platelets-TAPs. To the best of our knowledge, thus far this terminology has not been employed by anyone. Platelets are the first to appear at the site of the inflammatory process that accompanies cancer development. Within the first few hours from the start of the colonization of cancer cells platelet-tumor aggregates are responsible for neutrophils recruitment, and further release a number of factors associated with tumor growth, metastasis and neoangiogenesis. On the other hand, it also has been indicated that factors delivered from platelets can induce a cytotoxic effect on the proliferating neoplastic cells, and even enhance apoptosis. Undoubtedly, TAPs' role seems to be more complex when compared to tumor associated neutrophils and macrophages, which do not allow for their division into TAP P1 and TAP P2, as in the case of TANs and TAMs. In this review we discuss the role of TAPs as an important element of tumor invasiveness and as a potentially new therapeutic target to prevent cancer development. Nevertheless, better exploring the interactions between platelets and tumor cells could help in the formulation of new therapeutic goals that support or improve the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Violetta Dymicka-Piekarska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Olga M. Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Justyna Zińczuk
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Ewa Kratz
- Department of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wrocław, Poland
| | - Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| |
Collapse
|
85
|
Marques P, de Vries F, Dekkers OM, van Furth WR, Korbonits M, Biermasz NR, Pereira AM. Pre-operative serum inflammation-based scores in patients with pituitary adenomas. Pituitary 2021; 24:334-350. [PMID: 33230695 DOI: 10.1007/s11102-020-01112-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Full blood count (FBC) and serum inflammation-based scores reflect systemic inflammation and predict outcomes in cancer, but little is known in pituitary adenomas (PAs). We aimed to characterise FBC and inflammation-based scores in PA patients and investigate their usefulness in predicting challenging disease course. METHODS We studied 424 PA patients first operated at our centre with available pre-operative biochemical data. Patients with infection, malignancies, autoimmune or haematological conditions were excluded. Inflammation-based scores studied: Neutrophil-to-Lymphocyte Ratio (NLR), Platelet-to-Lymphocyte Ratio (PLR), Lymphocyte-to-Monocyte Ratio (LMR), Systemic Immune-Inflammation Index (SII), Neutrophil-Platelet Score (NPS), Prognostic Nutrition Index (PNI), and Glasgow Prognostic Score (GPS). RESULTS Cushing's disease patients had more platelets, leucocytes, neutrophils and monocytes, and higher NLR, NPS and SII. Serum inflammation-based scores didn't differ among non-Cushing PA subtypes. The glucocorticoid excess severity influenced leucocyte, eosinophil, basophil and platelet counts, and GPS in Cushing's disease. Patients with functioning non-Cushing PAs with suprasellar extension, cavernous sinus invasion and hypopituitarism had GPS ≥ 1, while NPS ≥ 1 was associated with suprasellar extension. More invasive and difficult to treat corticotrophinomas were associated with fewer platelets pre-operatively (< 299.5 × 109/L predicting multimodal treatment). Non-functioning PA patients who suffered apoplexy had more leucocytes, neutrophils and monocytes, higher GPS ≥ 1 and fewer platelets; re-operated cases had fewer lymphocytes, higher NLR and PLR. CONCLUSIONS Serum inflammation-based scores may predict invasive/refractory PAs: GPS and PNI in non-functioning and functioning non-Cushing PAs; NPS in functioning non-Cushing PAs; NLR and PLR in non-functioning PAs. Platelets < 299.5 × 109/L predict multimodal treatment in Cushing's disease. Further studies are needed to confirm these observations.
Collapse
Affiliation(s)
- Pedro Marques
- Department of Medicine, Division of Endocrinology and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Friso de Vries
- Department of Medicine, Division of Endocrinology and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Olaf M Dekkers
- Department of Medicine, Division of Endocrinology and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Wouter R van Furth
- Department of Neurosurgery and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Leiden, The Netherlands
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nienke R Biermasz
- Department of Medicine, Division of Endocrinology and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Alberto M Pereira
- Department of Medicine, Division of Endocrinology and Center for Endocrine Tumors Leiden (CETL), Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
86
|
Tanaka T, Asano T, Sano M, Takei J, Hosono H, Nanamiya R, Nakamura T, Yanaka M, Harada H, Fukui M, Suzuki H, Uchida K, Nakagawa T, Kato Y, Kaneko MK. Development of Monoclonal Antibody PMab-269 Against California Sea Lion Podoplanin. Monoclon Antib Immunodiagn Immunother 2021; 40:124-133. [PMID: 34042540 DOI: 10.1089/mab.2021.0011] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The development of protein-specific antibodies is essential for understanding a wide variety of biological phenomena. Parasitic and viral infections and cancers are known to occur within California sea lion (Zalophus californianus) populations. However, sensitive and specific monoclonal antibodies (mAbs) for the pathophysiological analysis of California sea lion tissues have not yet been developed. A type I transmembrane glycoprotein, podoplanin (PDPN), is a known diagnostic marker of lymphatic endothelial cells. We have previously developed several anti-PDPN mAbs in various mammalian species, with applications in flow cytometry, Western blotting, and immunohistochemistry. In this study, we established a novel mAb against California sea lion PDPN (seaPDPN), clone PMab-269 (mouse IgG1, kappa), using a Cell-Based Immunization and Screening method. PMab-269 is specifically detected in seaPDPN-overexpressed Chinese hamster ovary (CHO)-K1 cells using flow cytometry and Western blotting. Moreover, PMab-269 clearly identified pulmonary type I alveolar cells, renal podocytes, and colon lymphatic endothelial cells in California sea lion tissues using immunohistochemistry. These findings demonstrate the usefulness of PMab-269 for the pathophysiological analysis of lung, kidney, and lymphatic tissues of the California sea lion.
Collapse
Affiliation(s)
- Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masato Sano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Junko Takei
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | - Hideki Hosono
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Harada
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | | | - Hiroyoshi Suzuki
- Department of Pathology and Laboratory Medicine, Sendai Medical Center, Sendai, Japan
| | - Kazuyuki Uchida
- Laboratories of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Takayuki Nakagawa
- Laboratories of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan.,New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
87
|
Hong YM, Yoon KT, Cho M. Systemic immune-inflammation index predicts prognosis of sequential therapy with sorafenib and regorafenib in hepatocellular carcinoma. BMC Cancer 2021; 21:569. [PMID: 34006248 PMCID: PMC8130266 DOI: 10.1186/s12885-021-08124-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 03/29/2021] [Indexed: 12/26/2022] Open
Abstract
Background Regorafenib has shown promising results as a second-line therapy for patients with hepatocellular carcinoma (HCC) who progressed on sorafenib. Although there have been several data regarding the efficacy of sequential therapy with sorafenib and that of regorafenib in real-life, specific inflammation markers for predicting the prognosis have not been studied. This study aimed to investigate prognostic value of systemic inflammatory markers in patients with HCC who received sorafenib-regorafenib sequential therapy. Methods We retrospectively analyzed medical data of patients who received regorafenib for the treatment of HCC after sorafenib failure. Progression free survival (PFS) and overall survival (OS) were assessed using the Kaplan–Meier survival curves. Univariate and multivariate analyses were performed to analyze the factors associated with survival. Results A total of 58 patients who received at least one dose of regroafenib and fulfilled the eligibility criteria, good performance status (Eastern Cooperative Oncology Group [ECOG] 0–1) and preserved liver function (Child-Pugh-A), were included in the analysis. The median PFS was 3 months (95% confidence interval [CI] = 0.981–5.019) and the median OS was 8 months (95% CI = 5.761–10.239). Elevated systemic immune-inflammation index (SII ≥340) was independently associated with poor OS. In multivariate analysis, the SII (hazard ratio [HR] = 2.211, 95% CI = 1.089–4.489, P = 0.028) and alpha-fetoprotein (AFP) (HR = 2.750, 95% CI = 1.259–6.010, P = 0.011) were independent predictors of OS. Conclusion Elevated SII is associated with poor OS in patients with HCC who received sequential therapy with sorafenib and regorafenib. In addition, when selecting a treatment strategy, the SII can be used in combination with the AFP level as a promising prognostic tool for HCC.
Collapse
Affiliation(s)
- Young Mi Hong
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea
| | - K T Yoon
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea
| | - Mong Cho
- Department of Internal Medicine, Liver Center, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, 20 Geumo-ro, Gyeongnam, 50612, Yangsan, South Korea.
| |
Collapse
|
88
|
Shah S, Karathanasi A, Revythis A, Ioannidou E, Boussios S. Cancer-Associated Thrombosis: A New Light on an Old Story. Diseases 2021; 9:34. [PMID: 34064390 PMCID: PMC8161803 DOI: 10.3390/diseases9020034] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/08/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer-associated thrombosis (CAT) is a rising and significant phenomenon, becoming the second leading cause of death in cancer patients. Pathophysiology of CAT differs from thrombosis in the non-cancer population. There are additional risk factors for thrombosis specific to cancer including cancer type, histology, and treatment, such as chemotherapy. Recently developed scoring systems use these risk factors to stratify the degree of risk and encourage thromboprophylaxis in intermediate- to high-risk patients. Anticoagulation is safely used for prophylaxis and treatment of CAT. Both of these have largely been with low-molecular-weight heparin (LMWH), rather than the vitamin K antagonist (VKA); however, there has been increasing evidence for direct oral anticoagulant (DOAC) use. Consequently, international guidelines have also adapted to recommend the role of DOACs in CAT. Using DOACs is a turning point for CAT, but further research is warranted for their long-term risk profile. This review will discuss mechanisms, risk factors, prophylaxis and management of CAT, including both LMWH and DOACs. There will also be a comparison of current international guidelines and how they reflect the growing evidence base.
Collapse
Affiliation(s)
- Sidrah Shah
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.S.); (A.K.); (A.R.)
| | - Afroditi Karathanasi
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.S.); (A.K.); (A.R.)
| | - Antonios Revythis
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.S.); (A.K.); (A.R.)
| | - Evangelia Ioannidou
- Department of Paediatrics and Child Health, West Suffolk Hospital NHS Foundation Trust, Hardwick Lane, Bury St Edmunds IP33 2QZ, UK;
| | - Stergios Boussios
- Department of Hematology/Medical Oncology, Medway NHS Foundation Trust, Windmill Road, Gillingham ME7 5NY, UK; (S.S.); (A.K.); (A.R.)
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, London SE1 9RT, UK
- AELIA Organization, 9th Km Thessaloniki-Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
89
|
Winarno GNA, Pasaribu M, Susanto H, Nisa AS, Harsono AB, Yuseran H, Suardi D, Trianasari N. The Platelet to Lymphocyte and Neutrophil to Lymphocyte Ratios in Predicting Response to Platinum-based Chemotherapy for Epithelial Ovarian Cancer. Asian Pac J Cancer Prev 2021; 22:1561-1566. [PMID: 34048186 PMCID: PMC8408401 DOI: 10.31557/apjcp.2021.22.5.1561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Indexed: 11/25/2022] Open
Abstract
Objective: The patients with advanced-stage ovarian cancer have higher factors complicating surgery; thus, the best choice for them is surgery with chemotherapy with six cycles of adjuvant chemotherapy. Generally, chemotherapy can be evaluated in various ways, phsychal examination, radiology examination, and laboratory examination. This study aims is to examine if the measurement of the platelet/lymphocyte ratio (PLR) and neutrophil/lymphocyte ratio (NLR) can be used to predict a patient’s response to chemotherapy. Methods: Analytic observational study with a case-control design conducted in the Dr. Hasan Sadikin Hospital in Bandung from 2017 to 2018. This study used the medical record of ovarian cancer patients with post-surgery complete blood counts and histopathological reports. The sample size was determined based on the categorical test’s statistical calculation to obtain a total number of at minimal 90 samples. All the study subjects who had undergone complete chemotherapy were followed up for 6 months. Their response to chemotherapy was assessed with a clinical examination, ultrasonography, and a CA-125 blood test every 3 months. Results: In 2017–2018, 504 patients were diagnosed with ovarian cancer at the Dr. Hasan Sadikin Hospital in Bandung, Indonesia. After reassessment, 116 patients had stage I to III ovarian cancer and underwent cytoreduction followed by platinum chemotherapy. The age, cancer stage, and types of epithelial cells in the platinum-sensitive and platinum-resistant patients were characterized. There were significant differences between the two groups in age and cancer stage characteristics (p < 0.05). The increase in platelet/lymphocyte (p = 0.003) and neutrophil/lymphocyte ratios (p = 0.026) are associated with the increase in the response to platinum chemotherapy against epithelium-based cancers. Conclusion: A patient’s NLR and PLR are strongly associated with his response to chemotherapy.
Collapse
Affiliation(s)
| | - Marihot Pasaribu
- Department of Obstetrics and Gynaecology Medical Faculty Universitas Mulawarman/Abdul Wahab Sjahranie Hospital, Samarinda, Indonesia
| | - Herman Susanto
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Aisyah Shofiatun Nisa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Ali Budi Harsono
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Hariadi Yuseran
- Department of Obstetrics and Gynaecology Medical Faculty Universitas Mulawarman/ Ulin Hospital, Banjarmasin, Indonesia
| | - Dodi Suardi
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin Hospital, Bandung, Indonesia
| | - Nurvita Trianasari
- Telkom of Economics and Business School, Telkom University, Bandung, Indonesia
| |
Collapse
|
90
|
Antiplatelet Therapy Combined with Anastrozole Induces Features of Partial EMT in Breast Cancer Cells and Fails to Mitigate Breast-Cancer Induced Hypercoagulation. Int J Mol Sci 2021; 22:ijms22084153. [PMID: 33923802 PMCID: PMC8074114 DOI: 10.3390/ijms22084153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/24/2022] Open
Abstract
Thromboembolic complications are a leading cause of morbidity and mortality in cancer patients. Cancer patients often present with an increased risk for thrombosis including hypercoagulation, so the application of antiplatelet strategies to oncology warrants further investigation. This study investigated the effects of anastrozole and antiplatelet therapy (aspirin/clopidogrel cocktail or atopaxar) treatment on the tumour responses of luminal phenotype breast cancer cells and induced hypercoagulation. Ethical clearance was obtained (M150263). Blood was co-cultured with breast cancer cell lines (MCF7 and T47D) pre-treated with anastrozole and/or antiplatelet drugs for 24 h. Hypercoagulation was indicated by thrombin production and platelet activation (morphological and molecular). Gene expression associated with the epithelial-to-mesenchymal transition (EMT) was assessed in breast cancer cells, and secreted cytokines associated with tumour progression were evaluated. Data were analysed with the PAST3 software. Our findings showed that antiplatelet therapies (aspirin/clopidogrel cocktail and atopaxar) combined with anastrozole failed to prevent hypercoagulation and induced evidence of a partial EMT. Differences in tumour responses that modulate tumour aggression were noted between breast cancer cell lines, and this may be an important consideration in the clinical management of subphenotypes of luminal phenotype breast cancer. Further investigation is needed before this treatment modality (combined hormone and antiplatelet therapy) can be considered for managing tumour associated-thromboembolic disorder.
Collapse
|
91
|
Fabricius HÅ, Starzonek S, Lange T. The Role of Platelet Cell Surface P-Selectin for the Direct Platelet-Tumor Cell Contact During Metastasis Formation in Human Tumors. Front Oncol 2021; 11:642761. [PMID: 33791226 PMCID: PMC8006306 DOI: 10.3389/fonc.2021.642761] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
Mammalian platelets, devoid of nuclei, are the smallest cells in the blood stream. They are essential for hemostasis, but also transmit cell signals that are necessary for regenerative and generative processes such as inflammation, immunity and tissue repair. In particular, in malignancies they are also associated with cell proliferation, angiogenesis, and epithelial-mesenchymal transition. Platelets promote metastasis and resistance to anti-tumor treatment. However, fundamental principles of the interaction between them and target cells within tumors are complex and still quite obscure. When injected into animals or circulating in the blood of cancer patients, cancer cells ligate platelets in a timely manner closely related to platelet activation either by direct contact or by cell-derived substances or microvesicles. In this context, a large number of different surface molecules and transduction mechanisms have been identified, although the results are sometimes species-specific and not always valid to humans. In this mini-review, we briefly summarize the current knowledge on the role of the direct and indirect platelet-tumor interaction for single steps of the metastatic cascade and specifically focus on the functional role of P-selectin.
Collapse
Affiliation(s)
- Hans-Åke Fabricius
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Starzonek
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
92
|
The endoplasmic reticulum protein SEC22B interacts with NBEAL2 and is required for megakaryocyte α-granule biogenesis. Blood 2021; 136:715-725. [PMID: 32384141 DOI: 10.1182/blood.2019004276] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Studies of inherited platelet disorders have provided many insights into platelet development and function. Loss of function of neurobeachin-like 2 (NBEAL2) causes gray platelet syndrome (GPS), where the absence of platelet α-granules indicates NBEAL2 is required for their production by precursor megakaryocytes. The endoplasmic reticulum is a dynamic network that interacts with numerous intracellular vesicles and organelles and plays key roles in their development. The megakaryocyte endoplasmic reticulum is extensive, and in this study we investigated its role in the biogenesis of α-granules by focusing on the membrane-resident trafficking protein SEC22B. Coimmunoprecipitation (co-IP) experiments using tagged proteins expressed in human HEK293 and megakaryocytic immortalized megakaryocyte progenitor (imMKCL) cells established binding of NBEAL2 with SEC22B, and demonstrated that NBEAL2 can simultaneously bind SEC22B and P-selectin. NBEAL2-SEC22B binding was also observed for endogenous proteins in human megakaryocytes using co-IP, and immunofluorescence microscopy detected substantial overlap. SEC22B binding was localized to a region of NBEAL2 spanning amino acids 1798 to 1903, where 2 GPS-associated missense variants have been reported: E1833K and R1839C. NBEAL2 containing either variant did not bind SEC22B coexpressed in HEK293 cells. CRISPR/Cas9-mediated knockout of SEC22B in imMKCL cells resulted in decreased NBEAL2, but not vice versa. Loss of either SEC22B or NBEAL2 expression resulted in failure of α-granule production and reduced granule proteins in imMKCL cells. We conclude that SEC22B is required for α-granule biogenesis in megakaryocytes, and that interactions with SEC22B and P-selectin facilitate the essential role of NBEAL2 in granule development and cargo stability.
Collapse
|
93
|
Zhou C, Wang Y, Wang Y, Lei L, Ji MH, Zhou G, Xia H, Yang JJ. Predicting lung adenocarcinoma prognosis with a novel risk scoring based on platelet-related gene expression. Aging (Albany NY) 2021; 13:8706-8719. [PMID: 33619234 PMCID: PMC8034940 DOI: 10.18632/aging.202682] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 02/01/2021] [Indexed: 04/09/2023]
Abstract
Lung adenocarcinoma is the most common subtype of non-small cell lung cancer, and platelet receptor-related genes are related to its occurrence and progression. A new prognostic indicator based on platelet receptor-related genes was developed with multivariate COX analysis. Prognostic markers based on platelet-related risk score perform moderately in prognosis prediction. The functional annotation of this risk model in high-risk patients shows that the pathways related to cell cycle, glycolysis and platelet-derived related factors are rich. It is worth noting that somatic mutation analysis shows that TTN and MUC16 have higher mutation burdens in high-risk patients. Moreover, the differential genes of high- and low-risk groups are regulated by copy number variation and miRNA. And we provide a free online nomogram web tool based on clinical factors and the risk score (https://wsxzaq.shinyapps.io/wsxzaq_nomogram/). The score has been verified among three independent external cohorts (GSE13213, GSE68465 and GSE72094), and is still an independent risk factor for lung adenocarcinoma. In addition, among the other 6 cancers, the OS prognosis of high and low-risk groups of PRS is different (P < 0.05). Our research results have screened multiple platelet differential genes with clinical significance and constructed a meaningful prognostic risk score (PRS).
Collapse
Affiliation(s)
- Chengmao Zhou
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- School of Medicine, Southeast University, Nanjing 210009, China
| | - Yongsheng Wang
- Department of Respiratory Medicine, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Ying Wang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Lei Lei
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Mu-Huo Ji
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Guoren Zhou
- Jiangsu Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Institute of Cancer Research, Nanjing 210009, China
| | - Hongping Xia
- Department of Pathology, School of Basic Medical Sciences & Key Laboratory of Antibody Technique of National Health Commission & Jiangsu Antibody Drug Engineering Research Center, Nanjing Medical University, Nanjing 211166, China
- School of Medicine, Southeast University, Nanjing 210009, China
- Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
- School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
94
|
Geranpayehvaghei M, Dabirmanesh B, Khaledi M, Atabakhshi-Kashi M, Gao C, Taleb M, Zhang Y, Khajeh K, Nie G. Cancer-associated-platelet-inspired nanomedicines for cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1702. [PMID: 33538125 DOI: 10.1002/wnan.1702] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/09/2020] [Accepted: 01/08/2021] [Indexed: 01/03/2023]
Abstract
Platelets, with hemostasis and thrombosis activities, are one of the key components in the blood circulation. As a guard, they rapidly respond to any abnormal blood vessel injury signal and release their granules' contents, which induce their adhesion and aggregation on wound site for hemostasis. Recently, increasing evidence has indicated that platelets are critically involved in the growth and metastasis of cancer cells by releasing a variety of cytokines and chemokines to stimulate cancer cell proliferation and various angiogenic regulators to accelerate tumor angiogenesis. Platelets also secrete active transforming growth factor beta (TGF-β) to promote the epithelial-mesenchymal transition of cancer cells and their extravasation from primary site, and form microthrombus on the surface of cancer cells to protect them from immune attack and high-speed shear force in the circulation. Therefore, blocking platelet-cancer cell interaction may be an attractive strategy to treat primary tumor and/or prevent cancer metastasis. However, systemic inhibition or depletion of platelets brings risk of severe bleeding complication. Cancer-associated-platelets-targeted nanomedicines and biomimetic nanomedicines coated with platelet membrane can be used for targeted anticancer drug delivery, due to their natural targeting ability to tumor cells and platelets. In the current review, we first summarized the platelet mechanisms of action in physiological condition and their multiple roles in cancer progression and conventional antiplatelet therapeutics. We then highlighted the recent progress on the design and fabrication of cancer-associated-platelet-targeted nanomedicines and platelet membrane coating nanomedicines for cancer therapy. Finally, we discussed opportunities and challenges and offered our thoughts for the future development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Marzieh Geranpayehvaghei
- Faculty of Biological Sciences, Department of Nanobiotechnology, Tarbiat Modares University, Tehran, Iran.,CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Bahareh Dabirmanesh
- Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Khaledi
- Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Mona Atabakhshi-Kashi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Chao Gao
- College of Pharmaceutical Science, Jilin University, Changchun, China
| | - Mohammad Taleb
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Khosro Khajeh
- Faculty of Biological Sciences, Department of Nanobiotechnology, Tarbiat Modares University, Tehran, Iran.,Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.,GBA Research Innovation Institute for Nanotechnology, Guangdong, China
| |
Collapse
|
95
|
Vega-Mendoza D, Cañas-Linares A, Flores-Alcantar A, Espinosa-Neira R, Melchy-Perez E, Vera-Estrella R, Auvynet C, Rosenstein Y. CD43 (sialophorin) is involved in the induction of extracellular matrix remodeling and angiogenesis by lung cancer cells. J Cell Physiol 2021; 236:6643-6656. [PMID: 33533043 DOI: 10.1002/jcp.30308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/17/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022]
Abstract
Aberrant expression of CD43 in malignant tumors of nonhematopoietic origin such as those from lung, cervix, colon, and breast has been shown to correlate with poor prognosis, providing tumor cells with enhanced motility, anchorage-independent growth, and in vivo tumor size, while protecting the cells of NK lysis and apoptosis. To further characterize the role of CD43 in cell transformation, we tested whether interfering its expression modified the capacity of the A549 non-small cell lung cancer cells to secrete molecules contributing to malignancy. The proteomic analysis of the secretome of serum-starved A549 cells revealed that cells expressing normal levels of CD43 released significantly high levels of molecules involved in extracellular matrix organization, angiogenesis, platelet degranulation, collagen degradation, and inflammation, as compared to CD43 RNAi cells. This data reveals a novel and unexpected role for CD43 in lung cancer development, mainly in remodeling the tumor microenvironment.
Collapse
Affiliation(s)
- Daniela Vega-Mendoza
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Alicia Cañas-Linares
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Angel Flores-Alcantar
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Roberto Espinosa-Neira
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico.,División de Investigación Básica, Laboratorio de Epigenética del Cáncer, Instituto Nacional de Cancerología, Ciudad de México, Mexico
| | - Erika Melchy-Perez
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Rosario Vera-Estrella
- Departamento de Biología Molecular de Plantas, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Constance Auvynet
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| | - Yvonne Rosenstein
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
96
|
Wang D, Hu X, Xiao L, Long G, Yao L, Wang Z, Zhou L. Prognostic Nutritional Index and Systemic Immune-Inflammation Index Predict the Prognosis of Patients with HCC. J Gastrointest Surg 2021; 25:421-427. [PMID: 32026332 PMCID: PMC7904713 DOI: 10.1007/s11605-019-04492-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/23/2019] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Systemic nutrition and inflammation are the critical factors in cancer initiation, evolution, and progression. This study aimed to evaluate the prognostic value of the prognostic nutritional index (PNI) and systemic immune-inflammation index (SII) in hepatocellular carcinoma (HCC) patients who underwent liver resection. METHODS A total of 202 HCC patients met the criteria and were included in the study. The receiver operating characteristic (ROC) curve was used to calculate the optimal PNI and SII cutoff values. The relationship between PNI/SII and clinicopathologic parameters was analyzed. The effect of PNI and SII on recurrence-free survival (RFS) and overall survival (OS) was investigated by Kaplan-Meier curves and Cox proportional hazards models. RESULTS The areas under the ROC curve for PNI and SII were 0.64 and 0.58. The ideal preoperative PNI and SII cutoff values were 50.25 and 461.5, respectively. Multivariate Cox regression analysis identified that the PNI (P = 0.001) and tumor diameter (P = 0.018) were significant prognostic markers for RFS, and that the PNI (P = 0.049), SII (P = 0.039) and tumor diameter (P = 0.001) were significant prognostic markers for OS. The median RFS in the PNI-low and PNI-high groups was 13.5 months and 23 months (P = 0.001), and that in the SII-low and SII-high groups was 18 months and 15 months (P = 0.03), respectively. The median OS in the PNI-low and PNI-high groups was 24 months and 39 months (P = 0.001), and that in the SII-low and SII-high groups was 36 months and 22 months (P = 0.002), respectively. CONCLUSION Interestingly, we found that PNI and SII could be important prognostic parameters for HCC patients who under hepatectomy.
Collapse
Affiliation(s)
- Dong Wang
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Xi Hu
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Liang Xiao
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Guo Long
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Lei Yao
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - ZhiMing Wang
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - LeDu Zhou
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
97
|
Wang X, Liu B, Xu M, Jiang Y, Zhou J, Yang J, Gu H, Ruan C, Wu J, Zhao Y. Blocking podoplanin inhibits platelet activation and decreases cancer-associated venous thrombosis. Thromb Res 2021; 200:72-80. [PMID: 33548843 DOI: 10.1016/j.thromres.2021.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Patients with cancer are at a high risk of venous thromboembolism (VTE), studies have shown that high expression of podoplanin (PDPN) in tumors is associated with increased risk of VTE. METHODS Two human malignant cell lines (NCI-H226 and C8161) expressing high levels of PDPN were selected to explore the role of platelet in cancer-associated venous thrombosis in vitro and in vivo. Immunohistochemical staining using anti-PDPN antibody was performed in the pulmonary carcinoma patients. RESULTS Both NCI-H226 and C8161 cells expressing high PDPN triggered platelet activation via CLEC-2 in vitro, which was abrogated by an anti-PDPN antibody SZ-168. Furthermore, the in vivo study revealed that injection of CHO-PDPN or C8161 in two mouse model of venous thrombosis activated platelets, increased platelet counts and enhanced thrombosis. More importantly, PDPN-enhanced thrombosis was reduced in mice treated with SZ168. A total of 63.3% tumor specimens stained positive for PDPN. High PDPN expression was associated with an increased risk of VTE and poor prognosis. CONCLUSIONS PDPN expression in tumors induced platelet activation and was related to a high risk of VTE via platelet activation. SZ168 inhibited PDPN-induced platelet activation in vitro and decreased the incidence of VTE in mice.
Collapse
Affiliation(s)
- Xia Wang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China
| | - Biao Liu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China
| | - Mengqiao Xu
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Yizhi Jiang
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Jundong Zhou
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China
| | - Jun Yang
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China
| | - Haidi Gu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China
| | - Changgeng Ruan
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Jinchang Wu
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215006, Jiangsu, China; The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China.
| | - Yiming Zhao
- Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis of the Ministry of Health, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China; Collaborative Innovation Center of Hematology, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
98
|
Mijic S, Dabrosin C. Platelet Activation In Situ in Breasts at High Risk of Cancer: Relationship with Mammographic Density and Estradiol. J Clin Endocrinol Metab 2021; 106:485-500. [PMID: 33180937 DOI: 10.1210/clinem/dgaa820] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Indexed: 12/21/2022]
Abstract
CONTEXT High mammographic density in postmenopausal women is an independent risk factor for breast cancer by undetermined mechanisms. No preventive therapy for this risk group is available. Activated platelets release growth factors that modulate the microenvironment into a protumorigenic state. Estrogens may affect the risk of breast cancer and platelet function. Whether platelets are activated in situ in breast cancer or in normal breast tissue at high risk of breast cancer and the association to estradiol remains elusive. OBJECTIVE To investigate whether platelets are activated in situ in breast cancers and in dense breast tissue of postmenopausal women and explore correlations between estradiol, released platelet factors, and inflammatory proteins. SETTING AND DESIGN Sampling of in vivo proteins was performed using microdialysis in a total of 71 women: 10 with breast cancer, 42 healthy postmenopausal women with different breast densities, and 19 premenopausal women. RESULTS Our data demonstrate increased levels of coagulation factors in dense breast tissue similar to that found in breast cancers, indicating excessive platelet activation. Premenopausal breasts exhibited similar levels of coagulation factors as postmenopausal dense breasts. Out of 13 coagulations factors that were upregulated in dense breasts, 5 exhibited significant correlations with estradiol, both locally in the breast and systemically. In breast tissue, positive correlations between coagulation factors and key inflammatory proteins and matrix metalloproteinases were detected. CONCLUSIONS Breast density, not estradiol, is the major determinant of local platelet activation. Inactivation of platelets may be a therapeutic strategy for cancer prevention in postmenopausal women with dense breasts.
Collapse
Affiliation(s)
- Sofija Mijic
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Charlotta Dabrosin
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
99
|
Lee SY, Lee SI, Min BW, Oh SC. Prognostic implication of systemic inflammatory markers in young patients with resectable colorectal cancer. Ann Surg Treat Res 2021; 100:25-32. [PMID: 33457394 PMCID: PMC7791188 DOI: 10.4174/astr.2021.100.1.25] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 12/27/2022] Open
Abstract
PURPOSE The prognosis of young colorectal cancer (CRC) patients has not fully been addressed. The prognostic significance of systemic inflammatory markers was examined in those patients. METHODS A total of 965 patients with resectable CRC were divided into young (≤ 50 years, n = 101) and old groups (> 51 years, n = 864). Neutrophil-to-lymphocyte ratio (NLR) > 5, derived NLR (dNLR) > 3, lymphocyte-to-monocyte ratio (LMR) < 2, platelet-to-lymphocyte ratio (PLR) > 150, and prognostic nutritional index (PNI) < 45 were analyzed for prognosis. Overall survival (OS) and progression-free survival (PFS) were compared using the log-rank test. A multivariate analysis was performed using a Cox proportional hazards regression model. RESULTS In the young group, NLR > 5, LMR < 2, and PNI < 45 were significantly associated with OS with univariate analyses. dNLR > 3 and those markers showed significance for PFS. LMR < 2 was a significant marker for poor PFS (hazard ratio [HR], 5.81; P = 0.020) in the multivariate analysis. In the old group, all inflammatory markers were significantly associated with OS and PFS with univariate analyses. LMR < 2 (HR, 2.66; P = 0.016) and PNI < 45 (HR, 2.14; P = 0.016) were independently associated with OS in multivariate analyses. PLR > 150 (HR, 1.45; P = 0.036) and PNI < 45 (HR, 1.73; P = 0.002) were significant markers for PFS. CONCLUSION Systemic inflammation might be one of biologic factors that influence on prognosis of young CRC.
Collapse
Affiliation(s)
- Suk-young Lee
- Division of Hemato-Oncology, Department of Internal Medicine, Wonkwang University School of Medicine, Gunpo, Korea
| | - Sun Il Lee
- Department of Surgery, Korea University College of Medicine, Seoul, Korea
| | - Byung-Wook Min
- Department of Surgery, Korea University College of Medicine, Seoul, Korea
| | - Sang Cheul Oh
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| |
Collapse
|
100
|
Brady L, Hayes B, Sheill G, Baird AM, Guinan E, Stanfill B, Vlajnic T, Casey O, Murphy V, Greene J, Allott EH, Hussey J, Cahill F, Van Hemelrijck M, Peat N, Mucci L, Cunningham M, Grogan L, Lynch T, Manecksha RP, McCaffrey J, O’Donnell D, Sheils O, O’Leary J, Rudman S, McDermott R, Finn S. Platelet cloaking of circulating tumour cells in patients with metastatic prostate cancer: Results from ExPeCT, a randomised controlled trial. PLoS One 2020; 15:e0243928. [PMID: 33338056 PMCID: PMC7748139 DOI: 10.1371/journal.pone.0243928] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 11/28/2020] [Indexed: 12/12/2022] Open
Abstract
Background Circulating tumour cells (CTCs) represent a morphologically distinct subset of cancer cells, which aid the metastatic spread. The ExPeCT trial aimed to examine the effectiveness of a structured exercise programme in modulating levels of CTCs and platelet cloaking in patients with metastatic prostate cancer. Methods Participants (n = 61) were randomised into either standard care (control) or exercise arms. Whole blood was collected for all participants at baseline (T0), three months (T3) and six months (T6), and analysed for the presence of CTCs, CTC clusters and platelet cloaking. CTC data was correlated with clinico-pathological information. Results Changes in CTC number were observed within group over time, however no significant difference in CTC number was observed between groups over time. Platelet cloaking was identified in 29.5% of participants. A positive correlation between CTC number and white cell count (WCC) was observed (p = 0.0001), in addition to a positive relationship between CTC clusters and PSA levels (p = 0.0393). Conclusion The presence of platelet cloaking has been observed in this patient population for the first time, in addition to a significant correlation between CTC number and WCC. Trial registration ClincalTrials.gov identifier NCT02453139.
Collapse
Affiliation(s)
- Lauren Brady
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Brian Hayes
- Department of Histopathology, Cork University Hospital, Cork, Ireland
- Department of Pathology, University College Cork, Cork, Ireland
| | - Gráinne Sheill
- Discipline of Physiotherapy, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Anne-Marie Baird
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Emer Guinan
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Bryan Stanfill
- Pacific Northwest National Laboratory, Richland, Washington, United States of America
| | - Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | | | | - John Greene
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Emma H. Allott
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Centre for Cancer Research and Cell Biology, Queen’s University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Juliette Hussey
- Discipline of Physiotherapy, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Fidelma Cahill
- King’s College London, School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology (TOUR), London, United Kingdom
| | - Mieke Van Hemelrijck
- King’s College London, School of Cancer and Pharmaceutical Sciences, Translational Oncology and Urology (TOUR), London, United Kingdom
| | - Nicola Peat
- Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Lorelei Mucci
- Harvard T.H. Chan school of Public Health, Boston, Massachusetts, United States of America
| | - Moya Cunningham
- Department of Radiation Oncology, St Luke’s Hospital, Dublin, Ireland
| | - Liam Grogan
- Department of Oncology, Beaumont Hospital, Dublin, Ireland
| | - Thomas Lynch
- Department of Urology, St James’s Hospital, Dublin, Ireland
| | - Rustom P. Manecksha
- Department of Urology, St James’s Hospital, Dublin, Ireland
- Department of Surgery, Trinity College Dublin, Dublin, Ireland
| | - John McCaffrey
- Department of Oncology, Mater Misericordiae Hospital, Dublin, Ireland
| | | | - Orla Sheils
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - John O’Leary
- Department of Histopathology, St James’s Hospital, Dublin, Ireland
| | - Sarah Rudman
- Guy’s and St Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Ray McDermott
- Department of Oncology, Tallaght University Hospital, Dublin, Ireland
| | - Stephen Finn
- Department of Histopathology and Morbid Anatomy, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Cancer Trials Ireland, Dublin, Ireland
- Department of Histopathology, St James’s Hospital, Dublin, Ireland
- * E-mail:
| |
Collapse
|