1
|
Ciccioli M, Kim K, Khazan N, Khoury JD, Cooke MJ, Miller MC, O'Shannessy DJ, Pailhes-Jimenez AS, Moore RG. Identification of circulating tumor cells captured by the FDA-cleared Parsortix ® PC1 system from the peripheral blood of metastatic breast cancer patients using immunofluorescence and cytopathological evaluations. J Exp Clin Cancer Res 2024; 43:240. [PMID: 39169412 PMCID: PMC11337573 DOI: 10.1186/s13046-024-03149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/03/2024] [Indexed: 08/23/2024] Open
Abstract
Circulating Tumor Cells (CTCs) may serve as a non-invasive source of tumor material to investigate an individual's disease in real-time. The Parsortix® PC1 System, the first FDA-cleared medical device for the capture and harvest of CTCs from peripheral blood of metastatic breast cancer (MBC) patients for use in subsequent user-validated downstream analyses, enables the epitope-independent capture of CTCs with diverse phenotypes based on cell size and deformability. The aim of this study was to determine the proportion of MBC patients and self-declared female healthy volunteers (HVs) that had CTCs identified using immunofluorescence (IF) or Wright-Giemsa (WG) staining. Peripheral blood from 76 HVs and 76 MBC patients was processed on Parsortix® PC1 Systems. Harvested cells were cytospun onto a charged slide and immunofluorescently stained for identification of CTCs expressing epithelial markers. The IF slides were subsequently WG-stained and analyzed for CTC identification based on morphological features of malignant cells. All testing was performed by operators blinded to the clinical status of each subject. CTCs were identified on the IF slides in 45.3% (≥ 1) / 24.0% (≥ 5) of the MBC patients (range = 0 - 125, mean = 7) and in 6.9% (≥ 1) / 2.8% (≥ 5) of the HVs (range = 0 - 28, mean = 1). Among the MBC patients with ≥ 1 CTC, 70.6% had only CK + /EpCAM- CTCs, with none having EpCAM + /CK- CTCs. CTC clusters were identified in 56.0% of the CTC-positive patients. On the WG-stained slides, CTCs were identified in 42.9% (≥ 1) / 21.4% (≥ 5) of the MBC patients (range = 0 - 41, mean = 4) and 4.3% (≥ 1) / 2.9% (≥ 5) of the HVs (range = 0 - 14, mean = 0). This study demonstrated the ability of the Parsortix® PC1 System to capture and harvest CTCs from a significantly larger proportion of MBC patients compared to HVs when coupled with both IF and WG cytomorphological assessment. The presence of epithelial cells in subjects without diagnosed disease has been previously described, with their significance being unclear. Interestingly, a high proportion of the identified CTCs did not express EpCAM, highlighting the limitations of using EpCAM-based approaches.
Collapse
Affiliation(s)
| | - Kyukwang Kim
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Negar Khazan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Joseph D Khoury
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | - Richard G Moore
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
2
|
Qian J, Jiang Y, Hu H. Ginsenosides: an immunomodulator for the treatment of colorectal cancer. Front Pharmacol 2024; 15:1408993. [PMID: 38939839 PMCID: PMC11208871 DOI: 10.3389/fphar.2024.1408993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024] Open
Abstract
Ginsenosides, the primary bioactive ingredients derived from the root of Panax ginseng, are eagerly in demand for tumor patients as a complementary and alternative drug. Ginsenosides have increasingly become a "hot topic" in recent years due to their multifunctional role in treating colorectal cancer (CRC) and regulating tumor microenvironment (TME). Emerging experimental research on ginsenosides in the treatment and immune regulation of CRC has been published, while no review sums up its specific role in the CRC microenvironment. Therefore, this paper systematically introduces how ginsenosides affect the TME, specifically by enhancing immune response, inhibiting the activation of stromal cells, and altering the hallmarks of CRC cells. In addition, we discuss their impact on the physicochemical properties of the tumor microenvironment. Furthermore, we discuss the application of ginsenosides in clinical treatment as their efficacy in enhancing tumor patient immunity and prolonging survival. The future perspectives of ginsenoside as a complementary and alternative drug of CRC are also provided. This review hopes to open up a new horizon for the cancer treatment of Traditional Chinese Medicine monomers.
Collapse
Affiliation(s)
- Jianan Qian
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanyu Jiang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongyi Hu
- Department of Gastroenterology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Ahluwalia P, Ballur K, Leeman T, Vashisht A, Singh H, Omar N, Mondal AK, Vaibhav K, Baban B, Kolhe R. Incorporating Novel Technologies in Precision Oncology for Colorectal Cancer: Advancing Personalized Medicine. Cancers (Basel) 2024; 16:480. [PMID: 38339232 PMCID: PMC10854941 DOI: 10.3390/cancers16030480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most heterogeneous and deadly diseases, with a global incidence of 1.5 million cases per year. Genomics has revolutionized the clinical management of CRC by enabling comprehensive molecular profiling of cancer. However, a deeper understanding of the molecular factors is needed to identify new prognostic and predictive markers that can assist in designing more effective therapeutic regimens for the improved management of CRC. Recent breakthroughs in single-cell analysis have identified new cell subtypes that play a critical role in tumor progression and could serve as potential therapeutic targets. Spatial analysis of the transcriptome and proteome holds the key to unlocking pathogenic cellular interactions, while liquid biopsy profiling of molecular variables from serum holds great potential for monitoring therapy resistance. Furthermore, gene expression signatures from various pathways have emerged as promising prognostic indicators in colorectal cancer and have the potential to enhance the development of equitable medicine. The advancement of these technologies for identifying new markers, particularly in the domain of predictive and personalized medicine, has the potential to improve the management of patients with CRC. Further investigations utilizing similar methods could uncover molecular subtypes specific to emerging therapies, potentially strengthening the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Tiffanie Leeman
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kumar Vaibhav
- Department of Neurosurgery, Augusta University, Augusta, GA 30912, USA;
| | - Babak Baban
- Departments of Neurology and Surgery, Augusta University, Augusta, GA 30912, USA;
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| |
Collapse
|
4
|
Guo S, Huang J, Li G, Chen W, Li Z, Lei J. The role of extracellular vesicles in circulating tumor cell-mediated distant metastasis. Mol Cancer 2023; 22:193. [PMID: 38037077 PMCID: PMC10688140 DOI: 10.1186/s12943-023-01909-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023] Open
Abstract
Current research has demonstrated that extracellular vesicles (EVs) and circulating tumor cells (CTCs) are very closely related in the process of distant tumor metastasis. Primary tumors are shed and released into the bloodstream to form CTCs that are referred to as seeds to colonize and grow in soil-like distant target organs, while EVs of tumor and nontumor origin act as fertilizers in the process of tumor metastasis. There is no previous text that provides a comprehensive review of the role of EVs on CTCs during tumor metastasis. In this paper, we reviewed the mechanisms of EVs on CTCs during tumor metastasis, including the ability of EVs to enhance the shedding of CTCs, protect CTCs in circulation and determine the direction of CTC metastasis, thus affecting the distant metastasis of tumors.
Collapse
Affiliation(s)
- Siyin Guo
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
5
|
Kulkarni A, Bazou D, Santos-Martinez MJ. Bleeding and Thrombosis in Multiple Myeloma: Platelets as Key Players during Cell Interactions and Potential Use as Drug Delivery Systems. Int J Mol Sci 2023; 24:15855. [PMID: 37958838 PMCID: PMC10647631 DOI: 10.3390/ijms242115855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy originated in the bone marrow and characterized by unhindered plasma cell proliferation that results in several clinical manifestations. Although the main role of blood platelets lies in hemostasis and thrombosis, platelets also play a pivotal role in a number of other pathological conditions. Platelets are the less-explored components from the tumor microenvironment in MM. Although some studies have recently revealed that MM cells have the ability to activate platelets even in the premalignant stage, this phenomenon has not been widely investigated in MM. Moreover, thrombocytopenia, along with bleeding, is commonly observed in those patients. In this review, we discuss the hemostatic disturbances observed in MM patients and the dynamic interaction between platelets and myeloma cells, along with present and future potential avenues for the use of platelets for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Anushka Kulkarni
- The School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, The University of Dublin, D02 PN40 Dublin, Ireland;
| | - Despina Bazou
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland;
| | - Maria José Santos-Martinez
- The School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, The University of Dublin, D02 PN40 Dublin, Ireland;
- School of Medicine, Trinity College Dublin, D02 R590 Dublin, Ireland
- Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| |
Collapse
|
6
|
Santini D, Botticelli A, Galvano A, Iuliani M, Incorvaia L, Gristina V, Taffon C, Foderaro S, Paccagnella E, Simonetti S, Fazio F, Scagnoli S, Pomati G, Pantano F, Perrone G, De Falco E, Russo A, Spinelli GP. Network approach in liquidomics landscape. J Exp Clin Cancer Res 2023; 42:193. [PMID: 37542343 PMCID: PMC10401883 DOI: 10.1186/s13046-023-02743-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/27/2023] [Indexed: 08/06/2023] Open
Abstract
Tissue-based biopsy is the present main tool to explore the molecular landscape of cancer, but it also has many limits to be frequently executed, being too invasive with the risk of side effects. These limits and the ability of cancer to constantly evolve its genomic profile, have recently led to the need of a less invasive and more accurate alternative, such as liquid biopsy. By searching Circulating Tumor Cells and residues of their nucleic acids or other tumor products in body fluids, especially in blood, but also in urine, stools and saliva, liquid biopsy is becoming the future of clinical oncology. Despite the current lack of a standardization for its workflows, that makes it hard to be reproduced, liquid biopsy has already obtained promising results for cancer screening, diagnosis, prognosis, and risk of recurrence.Through a more accessible molecular profiling of tumors, it could become easier to identify biomarkers predictive of response to treatment, such as EGFR mutations in non-small cell lung cancer and KRAS mutations in colorectal cancer, or Microsatellite Instability and Mismatch Repair as predictive markers of pembrolizumab response.By monitoring circulating tumor DNA in longitudinal repeated sampling of blood we could also predict Minimal Residual Disease and the risk of recurrence in already radically resected patients.In this review we will discuss about the current knowledge of limitations and strengths of the different forms of liquid biopsies for its inclusion in normal cancer management, with a brief nod to their newest biomarkers and its future implications.
Collapse
Affiliation(s)
- Daniele Santini
- Oncologia Medica A, Policlinico Umberto 1, La Sapienza Università Di Roma, Rome, Italy
| | - Andrea Botticelli
- Oncologia Medica A, Policlinico Umberto 1, La Sapienza Università Di Roma, Rome, Italy
| | - Antonio Galvano
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Michele Iuliani
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Selcetta, Italy
| | - Lorena Incorvaia
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Valerio Gristina
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Chiara Taffon
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Simone Foderaro
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Selcetta, Italy
| | - Elisa Paccagnella
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.So Della Repubblica 79, 04100, Latina, Italy
| | - Sonia Simonetti
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Selcetta, Italy
| | - Federico Fazio
- UOC Oncologia Territoriale, Polo Pontino, La Sapienza Università Di Roma, Latina, Italy.
| | - Simone Scagnoli
- Oncologia Medica A, Policlinico Umberto 1, La Sapienza Università Di Roma, Rome, Italy
| | | | - Francesco Pantano
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Department of Medicine and Surgery, Università Campus Bio-Medico Di Roma, Selcetta, Italy
| | - Giuseppe Perrone
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico Di Roma, Rome, Italy
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University of Rome, C.So Della Repubblica 79, 04100, Latina, Italy
- Mediterranea Cardiocentro, 80122, Naples, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Gian Paolo Spinelli
- UOC Oncologia Territoriale, Polo Pontino, La Sapienza Università Di Roma, Latina, Italy
| |
Collapse
|
7
|
David P, Mittelstädt A, Kouhestani D, Anthuber A, Kahlert C, Sohn K, Weber GF. Current Applications of Liquid Biopsy in Gastrointestinal Cancer Disease-From Early Cancer Detection to Individualized Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071924. [PMID: 37046585 PMCID: PMC10093361 DOI: 10.3390/cancers15071924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Worldwide, gastrointestinal (GI) cancers account for a significant amount of cancer-related mortality. Tests that allow an early diagnosis could lead to an improvement in patient survival. Liquid biopsies (LBs) due to their non-invasive nature as well as low risk are the current focus of cancer research and could be a promising tool for early cancer detection. LB involves the sampling of any biological fluid (e.g., blood, urine, saliva) to enrich and analyze the tumor's biological material. LBs can detect tumor-associated components such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs). These components can reflect the status of the disease and can facilitate clinical decisions. LBs offer a unique and new way to assess cancers at all stages of treatment, from cancer screenings to prognosis to management of multidisciplinary therapies. In this review, we will provide insights into the current status of the various types of LBs enabling early detection and monitoring of GI cancers and their use in in vitro diagnostics.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Dina Kouhestani
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Kahlert
- Department of Surgery, Carl Gustav Carus University Hospital, 01307 Dresden, Germany
| | - Kai Sohn
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
8
|
Sturgess V, Azubuike UF, Tanner K. Vascular regulation of disseminated tumor cells during metastatic spread. BIOPHYSICS REVIEWS 2023; 4:011310. [PMID: 38510161 PMCID: PMC10903479 DOI: 10.1063/5.0106675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/17/2023] [Indexed: 03/22/2024]
Abstract
Cancer cells can travel to other organs via interconnected vascular systems to form new lesions in a process known as metastatic spread. Unfortunately, metastasis remains the leading cause of patient lethality. In recent years, it has been demonstrated that physical cues are just as important as chemical and genetic perturbations in driving changes in gene expression, cell motility, and survival. In this concise review, we focus on the physical cues that cancer cells experience as they migrate through the lymphatic and blood vascular networks. We also present an overview of steps that may facilitate organ specific metastasis.
Collapse
Affiliation(s)
- Victoria Sturgess
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Udochi F. Azubuike
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| | - Kandice Tanner
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Building 37, Room 2132, Bethesda MD 20892, USA
| |
Collapse
|
9
|
Shi Q, Ji T, Tang X, Guo W. The role of tumor-platelet interplay and micro tumor thrombi during hematogenous tumor metastasis. Cell Oncol (Dordr) 2023; 46:521-532. [PMID: 36652166 DOI: 10.1007/s13402-023-00773-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In addition to their pivotal roles in coagulation and thrombosis, platelets are crucial in tumor progression, with plenty of clinical and experimental data demonstrating that the interplay of platelets and tumor cells is essential for hematogenous tumor metastasis. After detach from primary sites, tumor cells intravasate into the blood circulation becoming circulating tumor cells and induce platelet activation, aggregation and encasement around tumor cells to form micro tumor thrombi, which create a permissive tumor microenvironment for metastasis. Platelets in micro tumor thrombi protect tumor cells from immune surveillance and anoikis (detachment-triggered apoptosis) through various pathways, which are significant for tumor cell survival in the bloodstream. Moreover, platelets can facilitate tumor metastasis by expediting epithelial-mesenchymal transition (EMT), adhesion to the endothelium, angiogenesis, tumor proliferation processes and platelet-derived microvesicle (PMV) formation. CONCLUSIONS Here, we provide a synopsis of the current understanding of the formation of micro tumor thrombi and the role of micro tumor thrombi in tumor hematogenous metastasis based on the tumor-platelet interplay. We also highlight potential therapeutic strategies targeting platelets for tumor treatment, including cancer-associated platelet-targeted nanomedicines.
Collapse
Affiliation(s)
- Qianyu Shi
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Tao Ji
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China.
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, 100044, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| |
Collapse
|
10
|
Ghose D, Swain S, Patra CN, Jena BR, Rao MEB. Advancement and Applications of Platelet-inspired Nanoparticles: A Paradigm for Cancer Targeting. Curr Pharm Biotechnol 2023; 24:213-237. [PMID: 35352648 DOI: 10.2174/1389201023666220329111920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/16/2021] [Accepted: 12/28/2021] [Indexed: 11/22/2022]
Abstract
Platelet-inspired nanoparticles have ignited the possibility of new opportunities for producing similar biological particulates, such as structural cellular and vesicular components, as well as various viral forms, to improve biocompatible features that could improve the nature of biocompatible elements and enhance therapeutic efficacy. The simplicity and more effortless adaptability of such biomimetic techniques uplift the delivery of the carriers laden with cellular structures, which has created varied opportunities and scope of merits like; prolongation in circulation and alleviating immunogenicity improvement of the site-specific active targeting. Platelet-inspired nanoparticles or medicines are the most recent nanotechnology-based drug targeting systems used mainly to treat blood-related disorders, tumors, and cancer. The present review encompasses the current approach of platelet-inspired nanoparticles or medicines that have boosted the scientific community from versatile fields to advance biomedical sciences. Surprisingly, this knowledge has streamlined to development of newer diagnostic methods, imaging techniques, and novel nanocarriers, which might further help in the treatment protocol of the various diseased conditions. The review primarily focuses on the novel advancements and recent patents in nanoscience and nanomedicine that could be streamlined in the future for the management of progressive cancers and tumor targeting. Rigorous technological advancements like biomimetic stem cells, pH-sensitive drug delivery of nanoparticles, DNA origami devices, virosomes, nano cells like exosomes mimicking nanovesicles, DNA nanorobots, microbots, etc., can be implemented effectively for target-specific drug delivery.
Collapse
Affiliation(s)
- Debashish Ghose
- Department of Pharmaceutics, Roland Institute of Pharmaceutical Sciences, Berhampur, 760 010, Biju Patnaik University of Technology, Rourkela, Odisha-769015, India
| | - Suryakanta Swain
- Department of Pharmacy, School of Health Sciences, The Assam Kaziranga University, Koraikhowa, NH-37, Jorhat, 785006, Assam, India
| | - Chinam Niranjan Patra
- Department of Pharmaceutics, Roland Institute of Pharmaceutical Sciences, Berhampur, 760 010, Biju Patnaik University of Technology, Rourkela, Odisha-769015, India
| | - Bikash Ranjan Jena
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, 752050, Odisha, India
| | - Muddana Eswara Bhanoji Rao
- Calcutta Institute of Pharmaceutical Technology and AHS, Banitabla, Uluberia, Howrah, 711316, West Bengal, India
| |
Collapse
|
11
|
Application of tumor-educated platelets as new fluid biopsy markers in various tumors. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:114-125. [PMID: 36284061 DOI: 10.1007/s12094-022-02937-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
The incidence of malignant tumors is increasing year by year. Early detection and diagnosis of malignant tumors can improve the prognosis of patients and prolong their life. Pathological biopsy is the current gold standard for diagnosis, but the results of pathological biopsy are affected by the sampling site and cannot fully reflect the nature of the disease. Moreover, the invasive nature of pathological biopsy limits repeated detection. Liquid biopsies are non-invasive and can be used for early detection and monitoring of tumors, which considered to represent a promising tool. Platelets make themselves to be one of the richest liquid biopsy sources by the capacity to take up proteins and nucleic acids and alter their megakaryocyte-derived transcripts and proteins in response to external signals, which are called tumor-educated platelets (TEPs). In this article, we will review the application of tumor-educated platelets in various malignancies (nasopharyngeal carcinoma, prostate cancer, lung cancer, glioblastoma, colorectal cancer, pancreas cancer, ovarian cancer, sarcoma, breast cancer and hepatocellular carcinoma) and provide theoretical basis for the research of TEPs in tumor diagnosis, monitoring and treatment.
Collapse
|
12
|
Wang H, Yin X, Ma K, Wang Y, Fang T, Zhang Y, Xue Y. Nomogram Based on Preoperative Fibrinogen and Systemic Immune-Inflammation Index Predicting Recurrence and Prognosis of Patients with Borrmann Type III Advanced Gastric Cancer. J Inflamm Res 2023; 16:1059-1075. [PMID: 36936348 PMCID: PMC10019083 DOI: 10.2147/jir.s404585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023] Open
Abstract
Background and Objectives The prognosis is known to differ significantly among advanced gastric cancer (AGC) with Borrmann type III. This study aimed to evaluate the prognosis of these patients more individually. Methods We selected 542 AGC patients with Borrmann type III. We used the receiver operating characteristic curve to analyze the cutoff values of inflammation indexes, and used Kaplan-Meier and Log rank tests to analyze recurrence-free survival (RFS) and overall survival (OS). The independent risk factors for recurrence and prognosis were analyzed by Cox proportional hazards regression model. The nomogram models were constructed by R studio. Results Patients with high preoperative fibrinogen (F) and systemic immune-inflammation index (SII) levels had worse RFS and OS and higher risk of postoperative locoregional recurrence, hematogenous metastasis and lymph node metastasis. F and SII can combine with different clinicopathological features (all P<0.05) to construct nomograms to predict 5-year recurrence and prognosis, which both were superior to pTNM stage alone. Conclusion The nomogram models based on F and SII can evaluate AGC with Borrmann type III postoperative recurrence and prognosis.
Collapse
Affiliation(s)
- Hao Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Xin Yin
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Keru Ma
- Department of Thoracic Surgery, Esophagus and Mediastinum, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yufei Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Tianyi Fang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yao Zhang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yingwei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
- Correspondence: Yingwei Xue, Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, People’s Republic of China, Tel +86-13304646901, Email
| |
Collapse
|
13
|
Pancreatic Cancer Cells Induce MicroRNA Deregulation in Platelets. Int J Mol Sci 2022; 23:ijms231911438. [PMID: 36232741 PMCID: PMC9569638 DOI: 10.3390/ijms231911438] [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: 09/02/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer is a pathology with a high mortality rate since it is detected at advanced stages, so the search for early-stage diagnostic biomarkers is essential. Liquid biopsies are currently being explored for this purpose and educated platelets are a good candidate, since they are known to present a bidirectional interaction with tumor cells. In this work, we analyzed the effects of platelets on cancer cells’ viability, as determined by MTT, migration using transwell assays, clonogenicity in soft agar and stemness by dilution assays and stem markers’ expression. We found that the co-culture of platelets and pancreatic cancer cells increased the proliferation and migration capacity of BXCP3 cells, augmented clonogenicity and induced higher levels of Nanog, Sox2 and Oct4 expression. As platelets can provide horizontal transfer of microRNAs, we also determined the differential expression of miRNAs in platelets obtained from a small cohort of pancreatic cancer patients and healthy subjects. We found clear differences in the expression of several miRNAs between platelets of patients with cancer healthy subjects. Moreover, when we analyzed microRNAs from the platelets of the pancreatic juice and blood derived from each of the cancer patients, interestingly we find differences between the blood- and pancreatic juice-derived platelets suggesting the presence of different subpopulations of platelets in cancer patients, which warrant further analysis.
Collapse
|
14
|
Corvigno S, Johnson AM, Wong KK, Cho MS, Afshar-Kharghan V, Menter DG, Sood AK. Novel Markers for Liquid Biopsies in Cancer Management: Circulating Platelets and Extracellular Vesicles. Mol Cancer Ther 2022; 21:1067-1075. [PMID: 35545008 DOI: 10.1158/1535-7163.mct-22-0087] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 02/03/2023]
Abstract
Although radiologic imaging and histologic assessment of tumor tissues are classic approaches for diagnosis and monitoring of treatment response, they have many limitations. These include challenges in distinguishing benign from malignant masses, difficult access to the tumor, high cost of the procedures, and tumor heterogeneity. In this setting, liquid biopsy has emerged as a potential alternative for both diagnostic and monitoring purposes. The approaches to liquid biopsy include cell-free DNA/circulating tumor DNA, long and micro noncoding RNAs, proteins/peptides, carbohydrates/lectins, lipids, and metabolites. Other approaches include detection and analysis of circulating tumor cells, extracellular vesicles, and tumor-activated platelets. Ultimately, reliable use of liquid biopsies requires bioinformatics and statistical integration of multiple datasets to achieve approval in a Clinical Laboratory Improvement Amendments setting. This review provides a balanced and critical assessment of recent discoveries regarding tumor-derived biomarkers in liquid biopsies along with the potential and pitfalls for cancer detection and longitudinal monitoring.
Collapse
Affiliation(s)
- Sara Corvigno
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anna Maria Johnson
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kwong-Kwok Wong
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Min Soon Cho
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David G Menter
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil K Sood
- Department of Gynecologic Oncology & Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
15
|
Zhao J, Ruan J, Lv G, Shan Q, Fan Z, Wang H, Du Y, Ling L. Cell membrane-based biomimetic nanosystems for advanced drug delivery in cancer therapy: A comprehensive review. Colloids Surf B Biointerfaces 2022; 215:112503. [PMID: 35429736 DOI: 10.1016/j.colsurfb.2022.112503] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/08/2022] [Accepted: 04/08/2022] [Indexed: 12/30/2022]
Abstract
Natural types of cells display distinct characteristics with homotypic targeting and extended circulation in the blood, which are worthy of being explored as promising drug delivery systems (DDSs) for cancer therapy. To enhance their delivery efficiency, these cells can be combined with therapeutic agents and artificial nanocarriers to construct the next generation of DDSs in the form of biomimetic nanomedicines. In this review, we present the recent advances in cell membrane-based DDSs (CDDSs) and their applications for efficient cancer therapy. Different sources of cell membranes are discussed, mainly including red blood cells (RBC), leukocytes, cancer cells, stem cells and hybrid cells. Moreover, the extraction methods used for obtaining such cells and the mechanism contributing to the functional action of these biomimetic CDDSs are explained. Finally, a future perspective is proposed to highlight the limitations of CDDSs and the possible resolutions toward clinical transformation of currently developed biomimetic chemotherapies.
Collapse
Affiliation(s)
- Jianing Zhao
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Jian Ruan
- Yantai Center for Food and Drug Control, Yantai 264005, China
| | - Guangyao Lv
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Qi Shan
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China
| | - Zhiping Fan
- Institute of BioPharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| | - Yuan Du
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| | - Longbing Ling
- Key Laboratory of Molecular Pharmacology and Drug Evaluation (Ministry of Education of China), School of Pharmacy, Yantai University, Yantai 264005, China.
| |
Collapse
|
16
|
Liu T, Wang X, Guo W, Shao F, Li Z, Zhou Y, Zhao Z, Xue L, Feng X, Li Y, Tan F, Zhang K, Xue Q, Gao S, Gao Y, He J. RNA Sequencing of Tumor-Educated Platelets Reveals a Three-Gene Diagnostic Signature in Esophageal Squamous Cell Carcinoma. Front Oncol 2022; 12:824354. [PMID: 35615147 PMCID: PMC9124963 DOI: 10.3389/fonc.2022.824354] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
There is no cost-effective, accurate, and non-invasive method for the detection of esophageal squamous cell carcinoma (ESCC) in clinical practice. We aimed to investigate the diagnostic potential of tumor-educated platelets in ESCC. In this study, seventy-one ESCC patients and eighty healthy individuals were enrolled and divided into a training cohort (23 patients and 27 healthy individuals) and a validation cohort (48 patients and 53 healthy individuals). Next-generation RNA sequencing was performed on platelets isolated from peripheral blood of all participants, and a support vector machine/leave-one-out cross validation (SVM/LOOCV) approach was used for binary classification. A diagnostic signature composed of ARID1A, GTF2H2, and PRKRIR discriminated ESCC patients from healthy individuals with 91.3% sensitivity and 85.2% specificity in the training cohort and 87.5% sensitivity and 81.1% specificity in the validation cohort. The AUC was 0.924 (95% CI, 0.845–0.956) and 0.893 (95% CI, 0.821–0.966), respectively, in the training cohort and validation cohort. This 3-gene platelet RNA signature could effectively discriminate ESCC from healthy control. Our data highlighted the potential of tumor-educated platelets for the noninvasive diagnosis of ESCC. Moreover, we found that keratin and collagen protein families and ECM-related pathways might be involved in tumor progression and metastasis of ESCC, which might provide insights to understand ESCC pathobiology and advance novel therapeutics.
Collapse
Affiliation(s)
- Tiejun Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Shao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Cancer Institute of the Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao, China
| | - Zitong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhihong Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoli Feng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yin Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Zhang
- Department of Medical Examination for Cancer Prevention, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yibo Gao, ; Jie He,
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yibo Gao, ; Jie He,
| |
Collapse
|
17
|
Wang YF, Yin X, Fang TY, Wang YM, Zhang L, Zhang XH, Zhang DX, Zhang Y, Wang XB, Wang H, Xue YW. Prognostic significance of serum inflammation indices for different tumor infiltrative pattern types of gastric cancer. World J Gastrointest Oncol 2022; 14:897-919. [PMID: 35582101 PMCID: PMC9048526 DOI: 10.4251/wjgo.v14.i4.897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/08/2021] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inflammatory indices are considered to be potential prognostic biomarkers for patients with gastric cancer (GC). However, there is no evidence defining the prognostic significance of inflammatory indices for GC with different tumor infiltrative pattern (INF) types.
AIM To evaluate the significance of inflammatory indices and INF types in predicting the prognosis of patients with GC.
METHODS A total of 962 patients who underwent radical gastrectomy were retrospectively selected for this study. Patients were categorized into the expansive growth type (INFa), the intermediate type (INFb), and the infiltrative growth type (INFc) groups. The cutoff values of inflammatory indices were analyzed by receiver operating characteristic curves. The Kaplan–Meier method and log-rank test were used to analyze overall survival (OS). The chi-square test was used to analyze the association between inflammatory indices and clinical characteristics. The independent risk factors for prognosis in each group were analyzed by univariate and multivariate analyses based on logistic regression. Nomogram models were constructed by R studio.
RESULTS The INFc group had the worst OS (P < 0.001). The systemic immune-inflammation index (P = 0.039) and metastatic lymph node ratio (mLNR) (P = 0.003) were independent risk factors for prognosis in the INFa group. The platelet-lymphocyte ratio (PLR) (P = 0.018), age (P = 0.026), body mass index (P = 0.003), and postsurgical tumor node metastasis (pTNM) stage (P < 0.001) were independent risk factors for prognosis in the INFb group. The PLR (P = 0.021), pTNM stage (P = 0.028), age (P = 0.021), and mLNR (P = 0.002) were independent risk factors for prognosis in the INFc group. The area under the curve of the nomogram model for predicting 5-year survival in the INFa group, INFb group, and INFc group was 0.787, 0.823, and 0.781, respectively.
CONCLUSION The outcome of different INF types GC patients could be assessed by nomograms based on different inflammatory indices and clinicopathologic features.
Collapse
Affiliation(s)
- Yu-Fei Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xin Yin
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Tian-Yi Fang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Yi-Min Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Lei Zhang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xing-Hai Zhang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Dao-Xu Zhang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Yao Zhang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Xi-Bo Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Hao Wang
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| | - Ying-Wei Xue
- Department of Gastroenterological Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang Province, China
| |
Collapse
|
18
|
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
|
19
|
Amekyeh H, Alkhader E, Sabra R, Billa N. Prospects of Curcumin Nanoformulations in Cancer Management. Molecules 2022; 27:361. [PMID: 35056675 PMCID: PMC8777756 DOI: 10.3390/molecules27020361] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/27/2021] [Accepted: 01/03/2022] [Indexed: 02/06/2023] Open
Abstract
There is increasing interest in the use of natural compounds with beneficial pharmacological effects for managing diseases. Curcumin (CUR) is a phytochemical that is reportedly effective against some cancers through its ability to regulate signaling pathways and protein expression in cancer development and progression. Unfortunately, its use is limited due to its hydrophobicity, low bioavailability, chemical instability, photodegradation, and fast metabolism. Nanoparticles (NPs) are drug delivery systems that can increase the bioavailability of hydrophobic drugs and improve drug targeting to cancer cells via different mechanisms and formulation techniques. In this review, we have discussed various CUR-NPs that have been evaluated for their potential use in treating cancers. Formulations reviewed include lipid, gold, zinc oxide, magnetic, polymeric, and silica NPs, as well as micelles, dendrimers, nanogels, cyclodextrin complexes, and liposomes, with an emphasis on their formulation and characteristics. CUR incorporation into the NPs enhanced its pharmaceutical and therapeutic significance with respect to solubility, absorption, bioavailability, stability, plasma half-life, targeted delivery, and anticancer effect. Our review shows that several CUR-NPs have promising anticancer activity; however, clinical reports on them are limited. We believe that clinical trials must be conducted on CUR-NPs to ensure their effective translation into clinical applications.
Collapse
Affiliation(s)
- Hilda Amekyeh
- Department of Pharmaceutics, School of Pharmacy, University of Health and Allied Sciences, Ho PMB 31, Ghana;
| | - Enas Alkhader
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan;
| | - Rayan Sabra
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Nashiru Billa
- Pharmaceutical Sciences Department, College of Pharmacy, QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
20
|
Zhang L, Zhu Y, Wei X, Chen X, Li Y, Zhu Y, Xia J, Huang Y, Huang Y, Wang J, Pang Z. Nanoplateletsomes restrain metastatic tumor formation through decoy and active targeting in a preclinical mouse model. Acta Pharm Sin B 2022; 12:3427-3447. [PMID: 35967283 PMCID: PMC9366539 DOI: 10.1016/j.apsb.2022.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/16/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022] Open
Abstract
Platelets buoy up cancer metastasis via arresting cancer cells, enhancing their adhesion, and facilitating their extravasation through the vasculature. When deprived of intracellular and granular contents, platelet decoys could prevent metastatic tumor formation. Inspired by these, we developed nanoplatesomes by fusing platelet membranes with lipid membranes (P-Lipo) to restrain metastatic tumor formation more efficiently. It was shown nanoplateletsomes bound with circulating tumor cells (CTC) efficiently, interfered with CTC arrest by vessel endothelial cells, CTC extravasation through endothelial layers, and epithelial-mesenchymal transition of tumor cells as nanodecoys. More importantly, in the mouse breast tumor metastasis model, nanoplateletsomes could decrease CTC survival in the blood and counteract metastatic tumor growth efficiently by inhibiting the inflammation and suppressing CTC escape. Therefore, nanoplatelesomes might usher in a new avenue to suppress lung metastasis.
Collapse
Affiliation(s)
- Longlong Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yuefei Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xunbin Wei
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Ying Zhu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yiheng Huang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Materia Medica, Academy of Chinese and Western Integrative Medicine, Fudan University, Shanghai 201203, China
- Corresponding authors.
| | - Zhiqing Pang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Corresponding authors.
| |
Collapse
|
21
|
Huang Y, Liu W. Cell membrane-engineered nanoparticles for cancer therapy. J Mater Chem B 2022; 10:7161-7172. [DOI: 10.1039/d2tb00709f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell-membrane-coated nanotechnology involves dressing the synthetic nanoparticles (NPs) with membrane derived from different types of cells to endow the NPs with the properties of a specific cell type and to further...
Collapse
|
22
|
Biomimetic platelet membrane-coated Nanoparticles for targeted therapy. Eur J Pharm Biopharm 2022; 172:1-15. [DOI: 10.1016/j.ejpb.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/18/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023]
|
23
|
Li G, Zhou X, Tian L, Meng G, Li B, Yu H, Li Y, Huo Z, Du L, Ma X, Xu B. Identification of aberrantly methylated-differentially expressed genes and potential agents for Ewing sarcoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1557. [PMID: 34790763 PMCID: PMC8576650 DOI: 10.21037/atm-21-4972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/13/2021] [Indexed: 11/06/2022]
Abstract
Background Human DNA methylation is a common epigenetic regulatory mechanism, and it plays a critical role in various diseases. However, the potential role of DNA methylation in Ewing sarcoma (ES) is not clear. This study aimed to explore the regulatory roles of DNA methylation in ES. Methods The microarray data of gene expression and methylation were downloaded from the Gene Expression Omnibus (GEO) database, and analyzed via GEO2R. Venn analysis was then applied to identify aberrantly methylated-differentially expressed genes (DEGs). Subsequently, function and pathway enrichment analysis was conducted, a protein-protein interaction (PPI) network was constructed, and hub genes were determined. Besides, a connectivity map (CMap) analysis was performed to screen bioactive compounds for ES treatment. Results A total of 135 hypomethylated high expression genes and 523 hypermethylated low expression genes were identified. The hypomethylated high expression genes were enriched in signal transduction and the apoptosis process. Meanwhile, hypermethylated low expression genes were related to DNA replication and transcription regulation. The PPI network analysis indicated C3, TF, and TCEB1 might serve as diagnostic and therapeutic targets of ES. Furthermore, CMap analysis revealed 6 chemicals as potential options for ES treatment. Conclusions The introduction of DNA methylation characteristics over DEGs is helpful to understand the pathogenesis of ES. The identified hub aberrantly methylated DEGs and chemicals might provide some novel insights on ES treatment.
Collapse
Affiliation(s)
- Guowang Li
- Graduate School of Tianjin Medical University, Tianjin, China.,Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| | - Xuan Zhou
- Department of Pediatrics, Haikou Hospital of The Maternal and Child Health, Haikou, China
| | - Lijun Tian
- Graduate School of Tianjin Medical University, Tianjin, China.,Department of Orthopedic, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Gedong Meng
- Graduate School of Tianjin Medical University, Tianjin, China.,Department of Spine Surgery, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Bo Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hao Yu
- Graduate School of Tianjin Medical University, Tianjin, China
| | - Yongjin Li
- Graduate School of Tianjin Medical University, Tianjin, China.,Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| | - Zhenxin Huo
- Graduate School of Tianjin Medical University, Tianjin, China.,Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| | - Lilong Du
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| | - Xinlong Ma
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| | - Baoshan Xu
- Department of Minimally Invasive Spine Surgery, Tianjin Hospital, Tianjin, China
| |
Collapse
|
24
|
Circulating Tumor Cell Clusters Are Cloaked with Platelets and Correlate with Poor Prognosis in Unresectable Pancreatic Cancer. Cancers (Basel) 2021; 13:cancers13215272. [PMID: 34771436 PMCID: PMC8582483 DOI: 10.3390/cancers13215272] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/13/2021] [Indexed: 12/24/2022] Open
Abstract
Circulating tumor cells (CTCs) are known to be heterogeneous and clustered with tumor-associated cells, such as macrophages, neutrophils, fibroblasts, and platelets. However, their molecular profile and clinical significance remain largely unknown. Thus, we aimed to perform a comprehensive gene expression analysis of single CTCs and CTC clusters in patients with pancreatic cancer and to identify their potential clinical relevance to provide personalized medicine. Epitope-independent, rapid (>3 mL of whole blood/min) isolation of single CTCs and CTC clusters was achieved from a prospective cohort of 16 patients with unresectable pancreatic cancer using a centrifugal microfluidic device. Forty-eight mRNA expressions of individual CTCs and CTC clusters were analyzed to identify pancreatic CTC phenotype. CTC clusters had a larger proportion of mesenchymal expression than single CTCs (p = 0.0004). The presence of CTC clusters positively correlated with poor prognosis (progression-free survival, p = 0.0159; overall survival, p = 0.0186). Furthermore, we found that most CTCs in these patients (90.7%) were cloaked with platelets and found the presence of a positive correlation between the increase in CTC clusters and rapid disease progression during follow-ups. Efficient CTC cluster isolation and analysis techniques will enhance the understanding of complex tumor metastasis processes and can facilitate personalized disease management.
Collapse
|
25
|
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
|
26
|
Zhang T, Yuan K, Wang Y, Xu M, Cai S, Chen C, Ma J. Identification of Candidate Biomarkers and Prognostic Analysis in Colorectal Cancer Liver Metastases. Front Oncol 2021; 11:652354. [PMID: 34422629 PMCID: PMC8371911 DOI: 10.3389/fonc.2021.652354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 07/19/2021] [Indexed: 02/02/2023] Open
Abstract
Background Colorectal cancer (CRC), one of the most common malignant tumors worldwide, has a high mortality rate, especially for patients with CRC liver metastasis (CLM). However, CLM pathogenesis remains unclear. Methods We integrated multiple cohort datasets and databases to clarify and verify potential key candidate biomarkers and signal transduction pathways in CLM. GEO2R, DAVID 6.8, ImageGP, STRING, UALCAN, ONCOMINE, THE HUMAN PROTEIN ATLAS, GEPIA 2.0, cBioPortal, TIMER 2.0, DRUGSURV, CRN, GSEA 4.0.3, FUNRICH 3.1.3 and R 4.0.3 were utilized in this study. Results Sixty-three pairs of matched colorectal primary cancer and liver metastatic gene expression profiles were screened from three gene expression profiles (GSE6988, GSE14297 and GSE81558). Thirty-one up-regulated genes and four down-regulated genes were identified from these three gene expression profiles and verified by another gene expression profiles (GSE 49355) and TCGA database. Two pathways (IGFBP-IGF signaling pathway and complement-coagulation cascade), eighteen key differentially expressed genes (DEGs), six hub genes (SPARCL1, CDH2, CP, HP, TF and SERPINA5) and two biomarkers (CDH2 and SPARCL1) with significantly prognostic values were screened by multi-omics data analysis and verified by Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) cohort. Conclusions In this study, we identified a robust set of potential candidate biomarkers in CLM, which would provide potential value for early diagnosis and prognosis, and would promote molecular targeting therapy for CRC and CLM.
Collapse
Affiliation(s)
- Tianhao Zhang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Kaitao Yuan
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingzhao Wang
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mingze Xu
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shirong Cai
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuangqi Chen
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinping Ma
- Division of Gastrointestinal Surgery Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
27
|
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
|
28
|
Chong W, Zhang Z, Luo R, Gu J, Lin J, Wei Q, Li B, Myers R, Lu-Yao G, Kelly WK, Wang C, Yang H. Integration of circulating tumor cell and neutrophil-lymphocyte ratio to identify high-risk metastatic castration-resistant prostate cancer patients. BMC Cancer 2021; 21:655. [PMID: 34078304 PMCID: PMC8170812 DOI: 10.1186/s12885-021-08405-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), and circulating tumor cells (CTCs) have been associated with survival in castration-resistant prostate cancer (CRPC). However, no study has examined the prognostic value of NLR and PLR in the context of CTCs. METHODS Baseline CTCs from mCRPC patients were enumerated using the CellSearch System. Baseline NLR and PLR values were calculated using the data from routine complete blood counts. The associations of CTC, NLR, and PLR values, individually and jointly, with progression-free survival (PFS) and overall survival (OS), were evaluated using Kaplan-Meier analysis, as well as univariate and multivariate Cox models. RESULTS CTCs were detected in 37 (58.7%) of 63 mCRPC patients, and among them, 16 (25.4%) had ≥5 CTCs. The presence of CTCs was significantly associated with a 4.02-fold increased risk for progression and a 3.72-fold increased risk of death during a median follow-up of 17.6 months. OS was shorter among patients with high levels of NLR or PLR than those with low levels (log-rank P = 0.023 and 0.077). Neither NLR nor PLR was individually associated with PFS. Among the 37 patients with detectable CTCs, those with a high NLR had significantly shorter OS (log-rank P = 0.024); however, among the 26 patients without CTCs, the OS difference between high- and low-NLR groups was not statistically significant. Compared to the patients with CTCs and low NLR, those with CTCs and high levels of NLR had a 3.79-fold risk of death (P = 0.036). This association remained significant after adjusting for covariates (P = 0.031). Combination analyses of CTC and PLR did not yield significant results. CONCLUSION Among patients with detectable CTCs, the use of NLR could further classify patients into different risk groups, suggesting a complementary role for NLR in CTC-based prognostic stratification in mCRPC.
Collapse
Affiliation(s)
- Weelic Chong
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Zhenchao Zhang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Rui Luo
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jian Gu
- Department of Epidemiology, MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jianqing Lin
- Department of Medicine, GW Cancer Center, George Washington University, Washington, DC, 20037, USA
| | - Qiang Wei
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37235, USA
| | - Bingshan Li
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37235, USA
| | - Ronald Myers
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Grace Lu-Yao
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - William Kevin Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Chun Wang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| | - Hushan Yang
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
29
|
Kong J, Yao C, Dong S, Wu S, Xu Y, Li K, Ji L, Shen Q, Zhang Q, Zhan R, Cui H, Zhou C, Niu H, Li G, Sun W, Zheng L. ICAM-1 Activates Platelets and Promotes Endothelial Permeability through VE-Cadherin after Insufficient Radiofrequency Ablation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002228. [PMID: 33643788 PMCID: PMC7887603 DOI: 10.1002/advs.202002228] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 11/06/2020] [Indexed: 06/02/2023]
Abstract
Radiofrequency ablation (RFA) for hepatocellular carcinoma (HCC) often leads to aggressive local recurrence and increased metastasis, and vascular integrity and platelets are implicated in tumor metastasis. However, whether interactions between endothelial cells and platelets induce endothelial permeability in HCC after insufficient RFA remains unclear. Here, significantly increased CD62P-positive platelets and sP-selectin in plasma are observed in HCC patients after RFA, and tumor-associated endothelial cells (TAECs) activate platelets and are susceptible to permeability after heat treatment in the presence of platelets in vitro. In addition, tumors exhibit enhanced vascular permeability after insufficient RFA in mice; heat treatment promotes platelets-induced endothelial permeability through vascular endothelial (VE)-cadherin, and ICAM-1 upregulation in TAECs after heat treatment results in platelet activation and increased endothelial permeability in vitro. Moreover, the binding interaction between upregulated ICAM-1 and Ezrin downregulates VE-cadherin expression. Furthermore, platelet depletion or ICAM-1 inhibition suppresses tumor growth and metastasis after insufficient RFA in an orthotopic tumor mouse model, and vascular permeability decreases in ICAM-1-/- mouse tumor after insufficient RFA. The findings suggest that ICAM-1 activates platelets and promotes endothelial permeability in TAECs through VE-cadherin after insufficient RFA, and anti-platelet and anti-ICAM-1 therapy can be used to prevent progression of HCC after insufficient RFA.
Collapse
Affiliation(s)
- Jian Kong
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Changyu Yao
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Shuying Dong
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Shilun Wu
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Yangkai Xu
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Ke Li
- Beijing Tiantan HospitalChina National Clinical Research Center for Neurological DiseasesAdvanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100050P. R. China
| | - Liang Ji
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Qiang Shen
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Qi Zhang
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Rui Zhan
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Hongtu Cui
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Changping Zhou
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
| | - Haigang Niu
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Guoming Li
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Wenbing Sun
- Department of Hepatobiliary SurgeryBeijing Chaoyang HospitalCapital Medical UniversityBeijing100043P. R. China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesPeking University Health Science CenterKey Laboratory of Molecular Cardiovascular Sciences of Ministry of EducationKey Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides of Ministry of HealthBeijing Key Laboratory of Cardiovascular Receptors ResearchBeijing100191P. R. China
- Beijing Tiantan HospitalChina National Clinical Research Center for Neurological DiseasesAdvanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijing100050P. R. China
| |
Collapse
|
30
|
Das C, Ghosh M, Mukhopadhyay M, Chatterjee S, Naskar B. Molecular profiling and utility of cell-free DNA in nonsmall carcinoma of the lung: Study in a tertiary care hospital. J Cancer Res Ther 2021; 17:1389-1396. [DOI: 10.4103/jcrt.jcrt_99_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
31
|
Vucenik I, Druzijanic A, Druzijanic N. Inositol Hexaphosphate (IP6) and Colon Cancer: From Concepts and First Experiments to Clinical Application. Molecules 2020; 25:E5931. [PMID: 33333775 PMCID: PMC7765177 DOI: 10.3390/molecules25245931] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/07/2020] [Accepted: 12/12/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple human health-beneficial effects have been related to highly phosphorylated inositol hexaphosphate (IP6). This naturally occurring carbohydrate and its parent compound, myo-inositol (Ins), are abundantly present in plants, particularly in certain high-fiber diets, but also in mammalian cells, where they regulate important cellular functions. However, the striking and broad-spectrum anticancer activity of IP6, consistently demonstrated in different experimental models, has been in a spotlight of the scientific community dealing with the nutrition and cancer during the last several decades. First experiments were performed in colon cancer 30 years ago. Since then, it has been shown that IP6 reduces cell proliferation, induces apoptosis and differentiation of malignant cells with reversion to normal phenotype, affecting several critical molecular targets. Enhanced immunity and antioxidant properties also contribute to the tumor cell destruction. Although Ins possesses a modest anticancer potential, the best anticancer results were obtained from the combination of IP6 + Ins. Here we review the first experimental steps in colon cancer, when concepts and hypotheses were put together almost without real knowledge and present clinical studies, that were initiated in colon cancer patients. Available as a dietary supplement, IP6 + Ins has been shown to enhance the anticancer effect of conventional chemotherapy, controls cancer metastases, and improves quality of life in cancer patients. Emerging clinical and still vast amount of experimental data suggest its role either as an adjuvant or as an "alternative" to current chemotherapy for cancer.
Collapse
Affiliation(s)
- Ivana Vucenik
- Department of Medical and Research Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pathology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Ana Druzijanic
- Department of Oral Medicine and Periodontology, School of Medicine, Dental Medicine, University of Split, 21000 Split, Croatia;
| | - Nikica Druzijanic
- Department of Surgery, University Hospital Split, School of Medicine, University of Split, 21000 Split, Croatia;
| |
Collapse
|
32
|
Wang S, Duan Y, Zhang Q, Komarla A, Gong H, Gao W, Zhang L. Drug Targeting via Platelet Membrane-Coated Nanoparticles. SMALL STRUCTURES 2020; 1:2000018. [PMID: 33817693 PMCID: PMC8011559 DOI: 10.1002/sstr.202000018] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Platelets possess distinct surface moieties responsible for modulating their adhesion to various disease-relevant substrates involving vascular damage, immune evasion, and pathogen interactions. Such broad biointerfacing capabilities of platelets have inspired the development of platelet-mimicking drug carriers that preferentially target drug payloads to disease sites for enhanced therapeutic efficacy. Among these carriers, platelet membrane-coated nanoparticles (denoted 'PNPs') made by cloaking synthetic substrates with the plasma membrane of platelets have emerged recently. Their 'top-down' design combines the functionalities of natural platelet membrane and the engineering flexibility of synthetic nanomaterials, which together create synergy for effective drug delivery and novel therapeutics. Herein, we review the recent progress of engineering PNPs with different structures for targeted drug delivery, focusing on three areas, including targeting injured blood vessels to treat vascular diseases, targeting cancer cells for cancer treatment and detection, and targeting drug-resistant bacteria to treat infectious diseases. Overall, current studies have established PNPs as versatile nanotherapeutics for drug targeting with strong potentials to improve the treatment of various diseases.
Collapse
Affiliation(s)
- Shuyan Wang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Yaou Duan
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Qiangzhe Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Anvita Komarla
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Hua Gong
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Weiwei Gao
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Departments of NanoEngineering, Chemical Engineering Program, and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
33
|
Hernández-Camarero P, Amezcua-Hernández V, Jiménez G, García MA, Marchal JA, Perán M. Clinical failure of nanoparticles in cancer: mimicking nature's solutions. Nanomedicine (Lond) 2020; 15:2311-2324. [PMID: 32969312 DOI: 10.2217/nnm-2020-0234] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The use of nanotechnology has become a promising approach in the treatment of cancer. However, most intravenously injected nanoparticles (NPs) do not effectively reach the tumor mass due to the biological barriers in the body. In an attempt to unify clinical criteria and basic research, we have collected the latest studies and described novel alternatives such as the use of NPs covered with cell membranes to increase NP delivery efficiency. Furthermore, we focus on the prospect of using the cell's natural messengers, exosomes, as vehicles to transport anti-cancer agents and we discuss the technical complications involved. Finally, we propose novel approaches to produce engineered exosomes which may overcome such technical limitations in order to achieve a proper anti-cancer nanotherapy.
Collapse
Affiliation(s)
- Pablo Hernández-Camarero
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas SN, Jaén E-23071, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| | - Víctor Amezcua-Hernández
- Hospital Universitario Virgen de las Nieves, Department Medical Oncology, Av. de las Fuerzas Armadas, 2, E-18014 Granada, Spain
| | - Gema Jiménez
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain
| | - María A García
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain.,Department of Biochemistry & Molecular Biology & Immunology, University of Granada, Granada, Spain
| | - Juan A Marchal
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain.,Instituto de Investigación Biosanitaria IBS.GRANADA, Granada, E-18071, Spain.,Department of Human Anatomy & Embryology, Faculty of Medicine, University of Granada, Avda. de la Investigación 11, Granada E-18016, Spain
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Campus de las Lagunillas SN, Jaén E-23071, Spain.,Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Spain
| |
Collapse
|
34
|
Zhu K, Li P, Mo Y, Wang J, Jiang X, Ge J, Huang W, Liu Y, Tang Y, Gong Z, Liao Q, Li X, Li G, Xiong W, Zeng Z, Yu J. Neutrophils: Accomplices in metastasis. Cancer Lett 2020; 492:11-20. [PMID: 32745581 DOI: 10.1016/j.canlet.2020.07.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/14/2020] [Accepted: 07/24/2020] [Indexed: 12/16/2022]
Abstract
Metastasis is a critical cause of treatment failure and death in patients with advanced malignancies. Tumor cells can leave the primary site and enter the bloodstream; these circulating tumor cells then colonize target organs by overcoming blood shear stress, evading immune surveillance, and silencing the offensive capabilities of immune cells, eventually forming metastatic foci. From leaving the primary focus to the completion of distant metastasis, malignant tumor cells are supported and/or antagonized by certain immune cells. In particular, it has been found that myeloid granulocytes play an important role in this process. This review therefore aims to comprehensively describe the significance of neutrophils in solid tumor metastasis in terms of their supporting role in initiating the invasion and migration of tumor cells and assisting the colonization of circulating tumor cells in distant target organs, with the hope of providing insight into and ideas for anti-tumor metastasis treatment of tumor patients.
Collapse
Affiliation(s)
- Kunjie Zhu
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Panchun Li
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongzhen Mo
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jie Wang
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xianjie Jiang
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Weilun Huang
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Liu
- Department of Plastic and Cosmetic Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanyan Tang
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhaojian Gong
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiaoling Li
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; NHC Key Laboratory of Carcinogenesis, and Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| | - Jianjun Yu
- Department of Head and Neck Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
35
|
Ren Q, Zhu P, Zhang H, Ye T, Liu D, Gong Z, Xia X. Identification and validation of stromal-tumor microenvironment-based subtypes tightly associated with PD-1/PD-L1 immunotherapy and outcomes in patients with gastric cancer. Cancer Cell Int 2020; 20:92. [PMID: 32226313 PMCID: PMC7092673 DOI: 10.1186/s12935-020-01173-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Immunotherapies targeting programmed cell death 1 (PD-1) and programmed death-ligand 1 (PD-L1) have been approved for gastric cancer (GC) patients. However, a large proportion of patients with T-cell-inflamed tumor microenvironment do not respond to the PD-1/PD-L1 blockade. The stromal component of the tumor microenvironment has been associated with immunotherapy. This study aims to explore the clinical significance of the non-immune cells in the tumor microenvironment and their potential as biomarkers for immunotherapy. Methods A total of 383 patients with GC from the Cancer Genome Atlas (TCGA) cohort, 300 patients with GC from the GSE62254 cohort in Gene Expression Omnibus (GEO) were included in the study. A stromal score was generated using the ESTIMATE algorithm, and the likelihood of response to PD-1/PD-L1 immunotherapy of GC patients was predicted using the TIDE algorithm. The prognostic value of the stromal score from GC cases was evaluated by the Kaplan–Meier method and Cox regression analysis. Gene set enrichment analysis (GSEA) was also conducted. Results The stromal score showed significant differences in different molecular subtypes and T stages. Multivariate analyses further confirmed that the stromal score was an independent indicator of overall survival (OS) in the two cohorts. The low stromal score group showed higher tumor mutation burden (TMB) and micro-satellite instability (MSI), and was more sensitive to immune checkpoint inhibitor according to the TIDE algorithm. Activation of the transforming growth factor and epithelial–mesenchymal transition were observed in the high stromal score subtype, which is associated with T-cell suppression, and may be responsible for resistance to PD-1/PD-L1 therapy. BPIFB2 was confirmed as a hub gene relevant to immunotherapy. Conclusion The stromal score was associated with cancer progression and molecular subtypes, and may serve as a novel biomarker for predicting the prognosis and response to immunotherapy in patients with GC.
Collapse
Affiliation(s)
- Qianqian Ren
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Peng Zhu
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, 430022 China
| | - Hui Zhang
- Department of Internal Medicine, Wuhan Hankou Hospital, Wuhan, 430011 China
| | - Tianhe Ye
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Dehan Liu
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Zhao Gong
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, 430022 China
| | - Xiangwen Xia
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| |
Collapse
|
36
|
Chen L, Kong X, Wang Z, Wang X, Fang Y, Wang J. Pre-treatment systemic immune-inflammation index is a useful prognostic indicator in patients with breast cancer undergoing neoadjuvant chemotherapy. J Cell Mol Med 2020; 24:2993-3021. [PMID: 31989747 PMCID: PMC7077539 DOI: 10.1111/jcmm.14934] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
The systemic immune‐inflammation index (SII = N × P/L) based on neutrophil (N), platelet (P) and lymphocyte (L) counts is used to predict the survival of patients with malignant tumours and can fully reflect the balance between host inflammatory and immune status. This study is conducted to explore the potential prognostic significance of SII in patients with breast cancer undergoing neoadjuvant chemotherapy (NACT). A total of 262 patients with breast cancer received NACT were enrolled in this study. According to the receiver operating characteristic curve, the optimal cut‐off value of SII was divided into two groups: low SII group (<602 × 109/L) and high SII group (≥602 × 109/L). The associations between breast cancer and clinicopathological variables by SII were determined by chi‐squared test or Fisher's exact test. The Kaplan‐Meier plots and log‐rank test were used to determine clinical outcomes of disease‐free survival (DFS) and overall survival (OS). The prognostic value of SII was analysed by univariate and multivariate Cox proportional hazards regression models. The toxicity of NACT was accessed by National Cancer Institute Common Toxicity Criteria (NCICTC). According to univariate and multivariate Cox regression survival analyses, the results showed that the value of SII had prognostic significance for DFS and OS. The patients with low SII value had longer DFS and OS than those with high SII value (31.11 vs 40.76 months, HR: 1.075, 95% CI: 0.718‐1.610, P = .006; 44.47 vs 53.68 months, HR: 1.051, 95% CI: 0.707‐1.564, P = .005, respectively). The incidence of DFS and OS in breast cancer patients with low SII value was higher than that in those patients with high SII value in 3‐, 5‐ and 10‐year rates. The common toxicities after NACT were haematological and gastrointestinal reaction, and there were no differences by SII for the assessment of side effects of neoadjuvant chemotherapy. Meanwhile, the results also proved that breast cancer patients with low SII value and high Miller and Payne grade (MPG) survived longer than those breast cancer with high SII value and low MPG grade. In patients without lymph vessel invasion, these breast cancer patients with low SII value had better prognosis and lower recurrence rates than those with high SII value. Pre‐treatment SII with the advantage of reproducible, convenient and non‐invasive was a useful prognostic indicator for breast cancer patients undergoing neoadjuvant chemotherapy and is a promising biomarker for breast cancer on treatment strategy decisions.
Collapse
Affiliation(s)
- Li Chen
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Zhong X, Zhang H, Zhu Y, Liang Y, Yuan Z, Li J, Li J, Li X, Jia Y, He T, Zhu J, Sun Y, Jiang W, Zhang H, Wang C, Ke Z. Circulating tumor cells in cancer patients: developments and clinical applications for immunotherapy. Mol Cancer 2020; 19:15. [PMID: 31980023 PMCID: PMC6982393 DOI: 10.1186/s12943-020-1141-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/16/2020] [Indexed: 02/08/2023] Open
Abstract
Cancer metastasis is the leading cause of cancer-related death. Circulating tumor cells (CTCs) are shed into the bloodstream from either primary or metastatic tumors during an intermediate stage of metastasis. In recent years, immunotherapy has also become an important focus of cancer research. Thus, to study the relationship between CTCs and immunotherapy is extremely necessary and valuable to improve the treatment of cancer. In this review, based on the advancements of CTC isolation technologies, we mainly discuss the clinical applications of CTCs in cancer immunotherapy and the related immune mechanisms of CTC formation. In order to fully understand CTC formation, sufficiently and completely understood molecular mechanism based on the different immune cells is critical. This understanding is a promising avenue for the development of effective immunotherapeutic strategies targeting CTCs.
Collapse
Affiliation(s)
- Xiaoming Zhong
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hangtian Zhang
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuqing Liang
- The Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, Commonwealth of Pennsylvania, USA
| | - Zhuolin Yuan
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiachen Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xin Li
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yifan Jia
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tian He
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiangyuan Zhu
- School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yu Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wengting Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Zhang
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Cheng Wang
- Division of Nephrology, Department of medicine, The Fifth Hospital of Sun Yat-sen University, Zhuhai, 519000, Guangdong, China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
38
|
Liu J, Ma F, Sun B, Cong Y, Xuan L, Wang Q, Wu S. Predictive Value of Lymphocyte-Related Blood Parameters at the Time Point of Lymphocyte Nadir During Radiotherapy in Breast Cancer. Onco Targets Ther 2020; 13:151-161. [PMID: 32021263 PMCID: PMC6955599 DOI: 10.2147/ott.s233244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 11/23/2019] [Indexed: 11/23/2022] Open
Abstract
Purpose Various reports found a relationship between lymphocyte-related blood parameters (LRBP), including absolute lymphocyte counts (ALC), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) and prognosis of breast cancer. Most of the studies focused on LRBP pre-treatment. Seldom have studies focused on LRBP during radiotherapy. We intended to perform a retrospective cohort study on the prognostic value of LRBP at the time point of lowest ALC during radiotherapy for breast cancer. Patients and methods A total of 158 female patients were included in radiotherapy group because of the strict limitation standards of complete routine blood test results at pre-treatment and pre-operation, and at least once a week during radiotherapy. Besides 221 patients, including the 158 patients of radiotherapy group, were adopted in pre-treatment group and pre-operation group. Results ALC and PLR at the time point of lowest ALC during radiotherapy are prognostic predictors of breast cancer, and lower ALC and higher PLR are independent significant predictors of poor DFS. Besides, lower ALC, higher NLR and higher PLR at both pre-treatment and pre-operation were found to be independent variables for predicting poor DFS. Conclusion LRBP at pre-treatment, pre-operation, and during radiotherapy may serve as predictors of outcomes of breast cancer.
Collapse
Affiliation(s)
- Jiannan Liu
- Academy of Military Medical Sciences, Beijing 100039, People's Republic of China.,Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| | - Feiyan Ma
- Baoding No.1 Central Hospital Affiliated with Chengde Medical College, Baoding City, Hebei Province 071000, People's Republic of China
| | - Bing Sun
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| | - Yang Cong
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| | - Liang Xuan
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| | - Qian Wang
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| | - Shikai Wu
- Department of Radiation Oncology, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of the PLA), Beijing 100071, People's Republic of China
| |
Collapse
|
39
|
Cacho-Díaz B, García-Botello DR, Wegman-Ostrosky T, Reyes-Soto G, Ortiz-Sánchez E, Herrera-Montalvo LA. Tumor microenvironment differences between primary tumor and brain metastases. J Transl Med 2020; 18:1. [PMID: 31900168 PMCID: PMC6941297 DOI: 10.1186/s12967-019-02189-8] [Citation(s) in RCA: 274] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
The present review aimed to discuss contemporary scientific literature involving differences between the tumor microenvironment (TME) in melanoma, lung cancer, and breast cancer in their primary site and TME in brain metastases (BM). TME plays a fundamental role in the behavior of cancer. In the process of carcinogenesis, cells such as fibroblasts, macrophages, endothelial cells, natural killer cells, and other cells can perpetuate and progress carcinogenesis via the secretion of molecules. Oxygen concentration, growth factors, and receptors in TME initiate angiogenesis and are examples of the importance of microenvironmental conditions in the performance of neoplastic cells. The most frequent malignant brain tumors are metastatic in origin and primarily originate from lung cancer, breast cancer, and melanoma. Metastatic cancer cells have to adhere to and penetrate the blood-brain barrier (BBB). After traversing BBB, these cells have to survive by producing various cytokines, chemokines, and mediators to modify their new TME. The microenvironment of these metastases is currently being studied owing to the discovery of new therapeutic targets. In these three types of tumors, treatment is more effective in the primary tumor than in BM due to several factors, including BBB. Understanding the differences in the characteristics of the microenvironment surrounding the primary tumor and their respective metastasis might help improve strategies to comprehend cancer.
Collapse
Affiliation(s)
- Bernardo Cacho-Díaz
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico.
| | - Donovan R García-Botello
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Talia Wegman-Ostrosky
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Gervith Reyes-Soto
- Neuro-oncology Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Elizabeth Ortiz-Sánchez
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico
| | - Luis Alonso Herrera-Montalvo
- Research Unit, Instituto Nacional de Cancerología, Av. San Fernando 22. Col. Sección XVI. Tlalpan, 14080, Mexico City, ZC, Mexico.
| |
Collapse
|
40
|
Abstract
Nanotechnology has made remarkable contributions to clinical oncology. Nanotherapeutics and diagnostic tools have distinctive characteristics which allow them superior abilities to deliver therapeutics and imaging agents for radiation oncology. Compared to solid biopsies and imaging, the analysis of circulating tumor cells (CTCs) offers a more rapid, real-time, and less invasive method to monitor the dynamic molecular profiles of tumors. The potential of CTCs to be translated as a novel cancer biomarker has been demonstrated in numerous clinical studies. This review will discuss clinical applications of nanomaterials in radiation oncology and the implication of CTCs in cancer detection and monitoring.
Collapse
Affiliation(s)
- Bo Sun
- Radiation Oncology, The University of North Carolina at Chapel Hill, 125 Mason Farm Road, Marsico 2236, Chapel Hill, NC 27599, USA
| | - C Tilden Hagan
- UNC/NCSU Joint Department of Biomedical Engineering, 125 Mason Farm Road, Marsico 2120, Chapel Hill, NC 27599, USA
| | - Joseph Caster
- Radiation Oncology, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Andrew Z Wang
- Radiation Oncology, The University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
41
|
Platelets and Hepatocellular Cancer: Bridging the Bench to the Clinics. Cancers (Basel) 2019; 11:cancers11101568. [PMID: 31618961 PMCID: PMC6826649 DOI: 10.3390/cancers11101568] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/10/2019] [Accepted: 10/14/2019] [Indexed: 02/06/2023] Open
Abstract
Growing interest is recently being focused on the role played by the platelets in favoring hepatocellular cancer (HCC) growth and dissemination. The present review reports in detail both the experimental and clinical evidence published on this topic. Several growth factors and angiogenic molecules specifically secreted by platelets are directly connected with tumor progression and neo-angiogenesis. Among them, we can list the platelet-derived growth factor, the vascular endothelial growth factor, the endothelial growth factor, and serotonin. Platelets are also involved in tumor spread, favoring endothelium permeabilization and tumor cells’ extravasation and survival in the bloodstream. From the bench to the clinics, all of these aspects were also investigated in clinical series, showing an evident correlation between platelet count and size of HCC, tumor biological behavior, metastatic spread, and overall survival rates. Moreover, a better understanding of the mechanisms involved in the platelet–tumor axis represents a paramount aspect for optimizing both current tumor treatment and development of new therapeutic strategies against HCC.
Collapse
|
42
|
Väyrynen JP, Väyrynen SA, Sirniö P, Minkkinen I, Klintrup K, Karhu T, Mäkelä J, Herzig KH, Karttunen TJ, Tuomisto A, Mäkinen MJ. Platelet count, aspirin use, and characteristics of host inflammatory responses in colorectal cancer. J Transl Med 2019; 17:199. [PMID: 31196200 PMCID: PMC6567577 DOI: 10.1186/s12967-019-1950-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/05/2019] [Indexed: 02/08/2023] Open
Abstract
Background Platelets not only contribute to hemostasis but also to the regulation of inflammatory reactions and cancer pathogenesis. We hypothesized that blood platelet count would be associated with systemic inflammation, the densities of tumor infiltrating immune cells, and survival in colorectal cancer (CRC), and these relationships could be altered by aspirin use. Methods We measured blood platelet count in a cohort of 356 CRC patients and analyzed its relationships with tumor and patient characteristics including aspirin use, markers of systemic inflammation (modified Glasgow Prognostic Score, mGPS; serum levels of CRP, albumin, and 13 cytokines), blood hemoglobin levels, five types of tumor infiltrating immune cells (CD3, CD8, FoxP3, Neutrophil elastase, mast cell tryptase), and survival. Results Platelet count inversely correlated with blood hemoglobin levels (p < 0.001) and positively correlated with serum levels of CRP and multiple cytokines including IL-1RA, IL-4, IL-6, IL-7, IL-8, IL-12, IFNγ, and PDGF-BB (p < 0.001 for all), while aspirin use was not associated with the levels of systemic inflammatory markers. High platelet count was also associated with high mGPS (p < 0.001) but did not show statistically significant multivariable adjusted associations with the densities of tumor infiltrating immune cells. Higher platelet counts were observed in higher tumor stage (p < 0.001), but platelet count or aspirin use were not associated with patient survival. Conclusions High platelet count is associated with systemic inflammation in CRC. This study could not demonstrate statistically significant associations between platelet count, aspirin use, and the densities of tumor infiltrating immune cells. Electronic supplementary material The online version of this article (10.1186/s12967-019-1950-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Juha P Väyrynen
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland. .,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland. .,Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA.
| | - Sara A Väyrynen
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland.,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Päivi Sirniö
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Ilkka Minkkinen
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Kai Klintrup
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Toni Karhu
- Research Unit of Biomedicine and Biocenter of Oulu, University of Oulu, POB 5000, 90014, Oulu, Finland.,Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Jyrki Mäkelä
- Research Unit of Surgery, Anesthesia and Intensive Care, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Surgery, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Karl-Heinz Herzig
- Research Unit of Biomedicine and Biocenter of Oulu, University of Oulu, POB 5000, 90014, Oulu, Finland.,Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland.,Department of Gastroenterology and Metabolism, Poznan University of Medical Sciences, ul. Szpitalna 27/33, 60-572, Poznan, Poland
| | - Tuomo J Karttunen
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Anne Tuomisto
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| | - Markus J Mäkinen
- Cancer and Translational Medicine Research Unit, University of Oulu, POB 5000, 90014, Oulu, Finland.,Department of Pathology, Oulu University Hospital and Medical Research Center Oulu, POB 21, 90029, Oulu, Finland
| |
Collapse
|
43
|
Abstract
Liquid biopsies have been considered the holy grail in achieving effective cancer management, with blood tests offering a minimally invasive, safe, and sensitive alternative or complementary approach for tissue biopsies. Currently, blood-based liquid biopsy measurements focus on the evaluation of biomarker types, including circulating tumor DNA, circulating tumor cells, extracellular vesicles (exosomes and oncosomes), and tumor-educated platelets (TEPs). Despite the potential of individual techniques, each has its own advantages and disadvantages. Here, we provide further insight into TEPs.
Collapse
|
44
|
Brehm MA, Windhorst S. New options of cancer treatment employing InsP 6. Biochem Pharmacol 2019; 163:206-214. [PMID: 30797871 DOI: 10.1016/j.bcp.2019.02.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 02/19/2019] [Indexed: 12/21/2022]
Abstract
Many mechanistic studies have been performed to analyze the cellular functions of the highly phosphorylated molecule inositol hexakisphosphate (InsP6) in health and disease. While the physiological intracellular functions are well described, the mechanism of potential pharmacological effects on cancer cell proliferation is still controversial. There are numerous studies demonstrating that a high InsP6 concentration (≥75 µM) inhibits growth of cancer cells in vitro and in vivo. Thus, there is no doubt that InsP6 exhibits anticancer activity but the mechanism underlying the cellular effects of extracellular InsP6 on cancer cells is far from being understood. In addition, studies on the inhibitory effect of InsP6 on cancer progression in animal models ignore aspects of its bioavailability. Here, we review and critically discuss the uptake mechanism and the intracellular involvement in signaling pathways of InsP6 in cancer cells. We take into account the controversial findings on InsP6 plasma concentration, which is a critical aspect of pharmacological accessibility of InsP6 for cancer treatment. Further, we discuss novel findings with respect to the effect of InsP6 on normal and immune cells as well as on platelet aggregate size. Our goal is to stimulate further mechanistic studies into novel directions considering previously disregarded aspects of InsP6. Only when we fully understand the mechanism underlying the anticancer activity of InsP6 novel and more efficient treatment options can be developed.
Collapse
Affiliation(s)
- Maria A Brehm
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany.
| |
Collapse
|
45
|
Thrombin Generation and Cancer: Contributors and Consequences. Cancers (Basel) 2019; 11:cancers11010100. [PMID: 30654498 PMCID: PMC6356447 DOI: 10.3390/cancers11010100] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
The high occurrence of cancer-associated thrombosis is associated with elevated thrombin generation. Tumour cells increase the potential for thrombin generation both directly, through the expression and release of procoagulant factors, and indirectly, through signals that activate other cell types (including platelets, leukocytes and erythrocytes). Furthermore, cancer treatments can worsen these effects. Coagulation factors, including tissue factor, and inhibitors of coagulation are altered and extracellular vesicles (EVs), which can promote and support thrombin generation, are released by tumour and other cells. Some phosphatidylserine-expressing platelet subsets and platelet-derived EVs provide the surface required for the assembly of coagulation factors essential for thrombin generation in vivo. This review will explore the causes of increased thrombin production in cancer, and the availability and utility of tests and biomarkers. Increased thrombin production not only increases blood coagulation, but also promotes tumour growth and metastasis and as a consequence, thrombin and its contributors present opportunities for treatment of cancer-associated thrombosis and cancer itself.
Collapse
|
46
|
Circulating tumor DNA – Current state of play and future perspectives. Pharmacol Res 2018; 136:35-44. [DOI: 10.1016/j.phrs.2018.08.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 08/20/2018] [Accepted: 08/20/2018] [Indexed: 12/15/2022]
|
47
|
Hernández-Camarero P, Jiménez G, López-Ruiz E, Barungi S, Marchal JA, Perán M. Revisiting the dynamic cancer stem cell model: Importance of tumour edges. Crit Rev Oncol Hematol 2018; 131:35-45. [PMID: 30293704 DOI: 10.1016/j.critrevonc.2018.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/22/2018] [Indexed: 02/07/2023] Open
Abstract
The lack of an effective treatment against cancer is not only due to its huge heterogeneity, but also to the fact that we don't have an answer to the question on how cancer originates. Among the proposed models to explain the development of cancer, the hierarchical model has been widely accepted. Nevertheless, this model fails to explain several experimental observations such as the cancer stem cells (CSCs) location inside a tumour or the differences between primary and metastatic tumours. Moreover, increasing evidence shows that the CSC phenotype is not a rigid state. Here, we present a critical review on the assumed tumour development models emphasizing the relevance of the dynamic and changing nature of cancer and the CSCs population in which the tumour microenvironment plays a crucial role and we propose a new model of tumour origin that could have an impact on new therapeutic strategies.
Collapse
Affiliation(s)
| | - Gema Jiménez
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E- 18016, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E- 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain
| | - Elena López-Ruiz
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E- 18016, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E- 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain
| | - Shivan Barungi
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain
| | - Juan Antonio Marchal
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E- 18016, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain; Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, E- 18016, Spain; Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, E-18071, Spain.
| | - Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain; Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, E- 18016, Spain; Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, Granada E-18100, Spain.
| |
Collapse
|
48
|
Deciphering Platelet Kinetics in Diagnostic and Prognostic Evaluation of Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2018; 2018:9142672. [PMID: 30050894 PMCID: PMC6040256 DOI: 10.1155/2018/9142672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
Liver pathophysiology can, directly and indirectly, impose morphological or biochemical abnormalities of the platelets. Conversely, platelets are also able to regulate the promitogenic and profibrotic signals on liver pathobiology. Platelet contribution to the liver pathophysiology is typically facilitated by the platelet-derived growth factors that are sequestered in different subsets of alpha and dense granules, and the release of these growth factors is synchronized according to the stage and type of liver disease or injury. Thus, platelets harbor clinically relevant information with potential diagnostic and prognostic implications in liver diseases. Hepatocellular carcinoma (HCC) largely influences the platelet kinetics, and a growing body of evidence has recognized its association with HCC occurrence or prognosis. This narrative review summarizes the progress made on implicating platelet as a diagnostic and prognostic tool for HCC; the review also dissects the contradictory results from earlier studies and reflects how combining platelet-based information may enable more reliable test for diagnostic and prognostic evaluation of HCC.
Collapse
|
49
|
Wang S, Yuan J, Yang J, Li N, Liu R, Luan J, Ye D. Advancement of platelet-inspired nanomedicine. Platelets 2018; 29:690-694. [PMID: 29883255 DOI: 10.1080/09537104.2018.1475633] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Shujun Wang
- Department of Blood Transfusion, Nanjing General Hospital of PLA, Nanjing, China
| | - Jun Yuan
- Department of Blood Transfusion, Nanjing General Hospital of PLA, Nanjing, China
| | - Jie Yang
- Department of Hematology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Na Li
- Department of Blood Transfusion, Nanjing General Hospital of PLA, Nanjing, China
| | - Ran Liu
- Department of Hematology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Jianfeng Luan
- Department of Blood Transfusion, Nanjing General Hospital of PLA, Nanjing, China
| | - Dong Ye
- Department of Blood Transfusion, Nanjing General Hospital of PLA, Nanjing, China
| |
Collapse
|
50
|
Luo CL, Xu ZG, Chen H, Ji J, Wang YH, Hu W, Wang K, Zhang WW, Yuan CH, Wang FB. LncRNAs and EGFRvIII sequestered in TEPs enable blood-based NSCLC diagnosis. Cancer Manag Res 2018; 10:1449-1459. [PMID: 29922089 PMCID: PMC5997181 DOI: 10.2147/cmar.s164227] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Tissue biopsy-based cancer diagnosis has limitations because of the fact that tumor tissues are in constant evolution and extremely heterogeneous. The current study was aimed to examine whether tumor-educated blood platelets (TEPs) might be a potential all-in-one source for blood-based cancer diagnostics to overcome the limitations of conventional cancer biopsy. Methods In the present study, we evaluated the expression pattern of MAGI2 antisense RNA 3 (MAGI2-AS3) and ZNFX1 antisense RNA 1 (ZFAS1) in both plasma and platelets of 101 non-small-cell lung cancer (NSCLC) patients. Receiver operating characteristic (ROC) curve was generated to evaluate their diagnostic potential. In addition, epidermal growth factor receptor (EGFR) mutations were detected in DNA and RNA samples of platelets for companion diagnostics. Results Our results showed that the levels of MAGI2-AS3 and ZFAS1 in both plasma and platelets of NSCLC patients were significantly downregulated than those in healthy controls. A positive correlation of long noncoding RNA expression was observed between platelets and plasma (r=0.738 for MAGI2-AS3, r=0.751 for ZFAS1, respectively). By ROC analysis, we found that molecular interrogation of MAGI2-AS3 and ZFAS1 in TEPs and plasma can offer valuable diagnostic performance for NSCLC patients (area under the ROC curve [AUC]MAGI2-AS3= 0.853/0.892, and AUCZFAS1=0.780/0.744 for diagnosing adenocarcinoma and squamous cell carcinoma cases from controls, respectively). Clinicopathologic characteristic analysis further revealed that MAGI2-AS3 level significantly correlated with tumor–node–metastasis (TNM) stage (p=0.001 in TEPs, p=0.003 in plasma), lymph-node metastasis (p=0.016 in TEPs, p=0.023 in plasma), and distant metastasis (p=0.045 in TEPs, p=0.045 in plasma), while ZFAS1 level was only correlated with TNM stage (p=0.005 in TEPs, p=0.044 in plasma). Furthermore, EGFRvIII RNA existed in both TEPs and plasma, but EGFR intracellular mutations cannot be detected in DNA of TEPs isolated from NSCLC. Conclusion Our data suggested that TEP is a promising source for NSCLC diagnosis and companion diagnostics.
Collapse
Affiliation(s)
- Chang-Liang Luo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhi-Gao Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hao Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jia Ji
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yu-Hui Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Wang
- Department of Laboratory Medicine, Hubei Cancer Hospital, Wuhan, China
| | - Wu-Wen Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chun-Hui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fu-Bing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|