1
|
Li J, Liu J, Yang S, Xia Y, Meng Q, Sun B, Liu Y, Zhao B, Jin J, Xu H, Wang L, Zhang P, Cheng Z. PD-L1 positive platelets mediate resistance to immune checkpoint inhibitors in patients with colorectal cancer. Cell Commun Signal 2025; 23:29. [PMID: 39815258 PMCID: PMC11737274 DOI: 10.1186/s12964-025-02034-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Programmed cell death ligand 1 (PD-L1) expression on immune cells is correlated with the efficacy of immune checkpoint inhibitor (ICI) therapy in various types of cancer. Platelets are important components of the tumour microenvironment (TME) and are widely involved in the development of many types of cancer including colorectal cancer (CRC). However, the role of PD-L1 positive platelets in ICI therapy for CRC remains unknown. We hypothesized that PD-L1 positive platelets trigger and sustain CRC immunosuppression. METHODS The functional depletion effects of PD-L1 positive platelets on TME and immune cells were measured via western blotting, immunofluorescence staining, qRT-PCR, ELISpot and flow cytometry. In vivo, CD274 knockout (KO), CD8a KO, platelet-specific KO (PF4-Cre-Hsp90b1flox/flox) mouse models and a subcutaneous tumour model treated with aspirin and PD-L1 mAb were established in C57BL/6 N mice. RESULTS We found that PD-L1 positive platelets are correlated with a poor prognosis, CD8 + T cell exhaustion and serve as a novel noninvasive biomarker for predicting immunotherapy efficacy in patients with CRC. The transfer of PD-L1 from tumour cells to platelets in the TME depends on direct cell contact via the fibronectin-1/GPIbα/integrin α5β1 pathway. In turn, platelets can also induce PD-L1 expression on cancer cells. Animal experiments revealed that antiplatelet pharmacological agents and genetic knockout of platelets potentiated the antitumour effect of the PD-L1 mAb treatment in a CD8 + T cell dependent manner. CONCLUSIONS Our data suggest that PD-L1 positive platelets suppress CD8 + T cell immunity. Clinical combination treatment with ICIs and antiplatelet agents may be an effective therapeutic strategy for treating CRC.
Collapse
Affiliation(s)
- Jiacheng Li
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Jia Liu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Shifeng Yang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Yu Xia
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Qingzhe Meng
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Biying Sun
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Yansong Liu
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Bin Zhao
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China
| | - Jiaqi Jin
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Lihong Wang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Pengxia Zhang
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| | - Zhuoxin Cheng
- Digestive Disease Center, The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province, 154000, China.
| |
Collapse
|
2
|
Drum DL, Jallorina AG, Wan LS, Chang VT, Lee-Wong MF. Non-Genetic Biomarkers in Merkel Cell Carcinoma: Prognostic Implications and Predictive Utility for Response to Anti-PD-(L)1 Immune Checkpoint Inhibitors. Exp Dermatol 2025; 34:e70030. [PMID: 39791602 DOI: 10.1111/exd.70030] [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: 05/31/2024] [Revised: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Merkel cell carcinoma (MCC) is a skin cancer that arises due to either Merkel cell polyomavirus infection (MCPyV) or ultraviolet (UV) radiation exposure, presenting primarily in the head and neck region of fair-skinned males. The recent success of PD-(L)1 immune checkpoint inhibitors (ICIs) in locally advanced/metastatic MCC, with an objective response rate (ORR) around 50% and improved survival, as a first-line treatment has moved ICIs to the forefront of therapy for MCC and generated interest in identifying biomarkers to predict clinical response. The MCC tumour microenvironment (TME) contains various components of the adaptive and innate immune system. These components can contribute to tumour immune escape through immunosuppression by preventing entrance of other immune cells or by aiding in the cytotoxic clearance of tumour cells. We aim to combine information from studies of baseline and on-treatment monitoring of the TME to help predict the success of ICIs in MCC. This review enhances the understanding of how CD8 T cells, γδ T cells and macrophages may impact predictions of response rates to ICIs in MCC patients. These immune cells are non-genetic biomarkers that can also be used to determine prognosis in MCC treatment.
Collapse
Affiliation(s)
- David L Drum
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Anika G Jallorina
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Leo S Wan
- Department of Medicine, West Virginia School of Osteopathic Medicine, Lewisburg, West Virginia, USA
| | - Victor T Chang
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey School of Medicine, Newark, New Jersey, USA
- Section of Hematology/Oncology, Veterans Administration New Jersey Health Care System, East Orange, New Jersey, USA
| | - Mary F Lee-Wong
- Department of Medicine and Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Allergy and Immunology, Maimonides Medical Center, Brooklyn, New York, USA
| |
Collapse
|
3
|
Chen Q, Zhai B, Li J, Wang H, Liu Z, Shi R, Wu H, Xu Y, Ji S. Systemic immune-inflammatory index predict short-term outcome in recurrent/metastatic and locally advanced cervical cancer patients treated with PD-1 inhibitor. Sci Rep 2024; 14:31528. [PMID: 39732889 DOI: 10.1038/s41598-024-82976-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
This study aims to assess the predictive value of certain markers of inflammation in patients with locally advanced or recurrent/metastatic cervical cancer who are undergoing treatment with anti-programmed death 1 (PD-1) therapy. A total of 105 patients with cervical cancer, who received treatment involving immunocheckpoint inhibitors (ICIs), were included in this retrospective study. We collected information on various peripheral blood indices, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), systemic immune-inflammation index (SII), and prognostic nutritional index (PNI). To determine the appropriate cutoff values for these inflammatory markers, we performed receiver operating characteristic curve (ROC) analysis. Progression-free survival (PFS) was estimated using the Kaplan-Meier method, and we conducted both univariate and multivariate Cox regression analyses to evaluate the prognostic value of these markers. Out of the 105 patients who received ICI treatment, the median progression-free survival (mPFS) was 19.0 months. We obtained the patients' clinical characteristics, such as age, pathological type, therapy regimen, Figo stage, NLR, PLR, LMR, SII, and PNI from their medical records. The optimal cutoff values for NLR, PLR, LMR, SII, and PNI were determined as 3.76, 218.1, 3.34, 1147.7, 43.75, respectively. In the univariate analysis, age, pathological type, therapy regimen, Figo stage, and LMR were not found to be associated with PFS. However, high NLR(P=0.001), high PLR(P<0.001), high SII(P<0.001), and low PNI (P=0.003)were all associated with shorter PFS. Multivariate analysis indicated that SII (P=0.017) was an independent risk factor for PFS. This study highlights the potential use of SII as a predictor of progression-free survival in cervical cancer patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Baoqian Zhai
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China
| | - Jingjing Li
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Hui Wang
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China
| | - Zhengcao Liu
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Runjun Shi
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China
| | - Haohao Wu
- Department of Radiotherapy Oncology, Yancheng City No.1 People's Hospital, The Fourth Affiliated Hospital of Nantong University, Yancheng, 224000, JiangSu Province, China.
| | - Yingying Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, 215001, JiangSu Province, China.
| | - Shengjun Ji
- Department of Radiotherapy & Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215001, JiangSu Province, China.
| |
Collapse
|
4
|
Cheng X, Meng F, Wang R, Liu S, Li Q, Chen B, Xi M. Prognostic value of immuno-inflammatory biomarkers in esophageal squamous cell carcinoma patients receiving immunotherapy combined with chemoradiotherapy and its association with immuno-genomic landscape. BMC Cancer 2024; 24:1518. [PMID: 39696104 DOI: 10.1186/s12885-024-13298-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The clinical significance of immuno-inflammatory indicators and the underlying biological basis in patients with esophageal squamous cell carcinoma (ESCC) who receive chemoradiotherapy (CRT) combined with immunotherapy remains unclear. This study aims to evaluate the prognostic value of immuno-inflammatory biomarkers, develop a prognostic model, and explore the underlying mechanisms. METHODS This study included 212 ESCC patients who received CRT and anti-PD-1 immunotherapy. Association between progression-free survival (PFS) and immuno-inflammatory biomarkers, including absolute lymphocyte count (ALC), neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), lymphocyte-monocyte ratio was analyzed. A nomogram was built based on the independent prognostic factors identified using multivariable Cox regression model. Pre-treatment tumor samples from 47 patients were collected for RNA sequencing to investigate the immune-related tumor microenvironment. RESULTS Patients experienced significant changes in immuno-inflammatory biomarkers during CRT, which gradually recovered after radiotherapy. Body mass index < 18.5 (HR, 1.85; P = 0.032), N3 stage (HR, 2.41; P = 0.002), high pre-CRT PLR (HR, 1.53; P = 0.037), low ALC nadir (HR, 1.84; P = 0.006), and high post-CRT NLR (HR, 2.12; P = 0.002) were independent prognostic factors for unfavorable PFS, which were incorporated into a nomogram with a concordance index of 0.70 (95% CI, 0.67-0.72). High-risk patients stratified by the nomogram had worse survival and were associated with lower levels of leukocyte and T cell activation, proliferation, and migration and less intratumoral immune cell infiltration. CONCLUSIONS Pre-CRT PLR, ALC nadir during CRT, and post-CRT NLR were significantly associated with PFS in patients with ESCC receiving CRT and immunotherapy. A nomogram model with good prognostic ability was developed.
Collapse
Affiliation(s)
- Xingyuan Cheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China
| | - Fanjun Meng
- Department of Radiation Oncology, Jieyang People's Hospital, Jieyang Affiliated Hospital, Sun Yat-sen University, Jieyang, China
| | - Ruixi Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shiliang Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China
| | - Qiaoqiao Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China
| | - Baoqing Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China
| | - Mian Xi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangzhou, China.
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, No.651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
5
|
Cheng LY, Su PJ, Kuo MC, Lin CT, Luo HL, Chou CC, Huang SY, Wu CC, Chen CH, Huang CC, Tsai KL, Yu-Li Su H. Combining serum inflammatory markers and clinical factors to predict survival in metastatic urothelial carcinoma patients treated with immune checkpoint inhibitors. Ther Adv Med Oncol 2024; 16:17588359241305091. [PMID: 39687055 PMCID: PMC11648016 DOI: 10.1177/17588359241305091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background Despite the revolutionary impact of immune checkpoint inhibitors (ICIs) on the treatment of metastatic urothelial carcinoma (mUC), the clinical utility of reliable prognostic biomarkers to foresee survival outcomes remains underexplored. Objectives The purpose of this study was to ascertain the prognostic significance of serum inflammatory markers in mUC patients undergoing ICI therapy. Design This is a retrospective, multicenter study. Methods Data were collected from two independent medical centers in Taiwan, encompassing a validation and a training cohort (TC). Patients with histopathologically confirmed urothelial carcinoma who received at least one cycle of ICI monotherapy were included. Serum inflammatory markers such as neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and systemic immune-inflammation index (SII) were calculated prior to ICI therapy. Statistical analyses involved the use of receiver operating characteristic (ROC) curves to determine optimal biomarker cutoffs and Cox proportional hazards models to evaluate the independent predictive capability of these markers. Results A total of 192 patients were enrolled. In the univariate analysis, serum markers such as NLR, PLR, SII, and Hb were significantly associated with overall survival (OS) in both the training and validation cohorts (VC). White blood cells, NLR, and SII demonstrated a robust correlation with progression-free survival across both cohorts. Multivariate analysis revealed that Eastern Cooperative Oncology Group performance status ⩾2 (p < 0.001), visceral metastasis (p < 0.001), leukocytosis (p < 0.001), Hb levels ⩾10 mg/dL (p = 0.008), and NLR ⩾5 (p = 0.032) as independent predictors of OS. A prognostic nomogram integrating these independent factors yielded a C-index for a 3-year OS of 0.769 in the TC and 0.657 in the VC. Conclusion Serum inflammatory markers, combined with clinicopathologic factors, provide a practical prognostic tool in mUC treatment with ICIs.
Collapse
Affiliation(s)
- Liang-Yun Cheng
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Po-Jung Su
- Division of Hematology–Oncology, Chang Gung Memorial Hospital at Linkou and Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Ming-Chun Kuo
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chang-Ting Lin
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hao-Lun Luo
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Chi Chou
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shih-Yu Huang
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Che Wu
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Hsu Chen
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chun-Chieh Huang
- Department of Radiation Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kai-Lung Tsai
- Department of Colorectal Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Harvey Yu-Li Su
- Division of Hematology–Oncology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, No. 123, Dapi Road, Niaosong District, Kaohsiung City 833, Taiwan
- Genomic and Proteomic Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
6
|
Ge Y, Liu X, Xu Y, Su Y, Li Y, Wang L. Combined systemic immune-inflammatory index and prognostic nutritional index predicts the efficacy and prognosis of ES-SCLC patients receiving PD-L1 inhibitors combined with first-line chemotherapy. Front Oncol 2024; 14:1485849. [PMID: 39697233 PMCID: PMC11652344 DOI: 10.3389/fonc.2024.1485849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Background There is a strong association between inflammation and the formation, progression, and metastasis of malignant tumors, according to earlier studies. Some composite inflammation-nutritional indicators, such as the systemic immune-inflammation index (SII) and the prognostic nutritional index (PNI), have a certain predictive effect on the prognosis of patients with small cell lung cancer (SCLC). However, the relationship between these indicators and the efficacy of immunotherapy in SCLC patients is still not well understood. Therefore, the purpose of this study was to explore how the pre-treatment SII-PNI score can predict the tumor response and prognosis of extensive-stage SCLC patients treated with PD-L1 inhibitors and first-line chemotherapy. Methods This research conducted a retrospective review of 70 ES- SCLC patients from December 2019 to January 2023. According to the SII-PNI score, all patients were categorized into three groups. Overall survival (OS) was assessed by implementing the Kaplan Meier and Cox regression models. In addition, we devised a nomogram and scrutinized its accuracy in prediction through receiver operating characteristic (ROC) curve analysis and visualized it by calibration plots. Subsequently, a risk classification system was established. Results Patients with higher SII-PNI scores exhibited notably poorer survival outcomes compared to their counterpart with low SII-PNI score (p=0.008), as well as poorer short-term curative effects (p=0.004). The results of the multivariate analysis revealed that the SII-PNI score (p=0.036) had an independent association with a less favorable OS. The nomogram has been demonstrated to be a reliable prognostic tool for ES-SCLC patients. A notable difference was identified between the two different levels of risk. Conclusion The baseline SII-PNI score can serve as a reliable prognostic indicator for ES-SCLC patients receiving immunotherapy. Higher SII-PNI scores imply a worse prognosis.
Collapse
Affiliation(s)
- Yi Ge
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Liu
- Department of Oncology, Luohe Central Hospital, Luohe, China
| | - Yishi Xu
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanwei Su
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixin Li
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
7
|
Kurma K, Eslami-S Z, Alix-Panabières C, Cayrefourcq L. Liquid biopsy: paving a new avenue for cancer research. Cell Adh Migr 2024; 18:1-26. [PMID: 39219215 PMCID: PMC11370957 DOI: 10.1080/19336918.2024.2395807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The current constraints associated with cancer diagnosis and molecular profiling, which rely on invasive tissue biopsies or clinical imaging, have spurred the emergence of the liquid biopsy field. Liquid biopsy involves the extraction of circulating tumor cells (CTCs), circulating free or circulating tumor DNA (cfDNA or ctDNA), circulating cell-free RNA (cfRNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from bodily fluid samples. Subsequently, these components undergo molecular characterization to identify biomarkers that are critical for early cancer detection, prognosis, therapeutic assessment, and post-treatment monitoring. These innovative biosources exhibit characteristics analogous to those of the primary tumor from which they originate or interact. This review comprehensively explores the diverse technologies and methodologies employed for processing these biosources, along with their principal clinical applications.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
8
|
Xie Q, Zhou J, He C, Xu Y, Tao F, Hu M. Unlocking the intricacies: Exploring the complex interplay between platelets and ovarian cancer. Crit Rev Oncol Hematol 2024; 202:104465. [PMID: 39097249 DOI: 10.1016/j.critrevonc.2024.104465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Ovarian cancer, an aggressive malignancy of the female reproductive tract, is frequently linked to an elevated risk of thrombotic events. This association is manifested by a pronounced rise in platelet counts and activation levels. Current research firmly supports the pivotal role of platelets in the oncogenic processes of ovarian cancer, influencing tumor cell proliferation and metastasis. Platelets influence these processes through direct interactions with tumor cells or by secreting cytokines and growth factors that enhance tumor growth, angiogenesis, and metastasis. This review aims to thoroughly dissect the interactions between platelets and ovarian cancer cells, emphasizing their combined role in tumor progression and associated thrombotic events. Additionally, it summarizes therapeutic strategies targeting platelet-cancer interface which show significant promise. Such approaches could not only be effective in managing the primary ovarian tumor but also play a pivotal role in preventing metastasis and attenuating thrombotic complications associated with ovarian cancer.
Collapse
Affiliation(s)
- Qianxin Xie
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Zhou
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaonan He
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Mengjiao Hu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
9
|
Guo L, Li J, Wang J, Chen X, Cai C, Zhou F, Xiong A. Prognostic role of dynamic changes in inflammatory indicators in patients with non-small cell lung cancer treated with immune checkpoint inhibitors-a retrospective cohort study. Transl Lung Cancer Res 2024; 13:1975-1987. [PMID: 39263031 PMCID: PMC11384502 DOI: 10.21037/tlcr-24-637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
Background Immune checkpoint inhibitors (ICIs) have become one of the standard treatments for non-small cell lung cancer (NSCLC) patients without driver mutations. However, a considerable proportion of patients suffer from severe immune side effects and fail to respond to ICIs. As effective biomarkers, programmed cell death ligand 1 (PD-L1) expression, microsatellite instability (MSI), the tumor mutation burden (TMB) and tumor-infiltrating lymphocytes (TILs) require invasive procedures that place heavy physical and psychological burdens on patients. This study aims to identify simple and effective markers to optimize patient selection through therapeutic decisions and outcome prediction. Methods This retrospective study comprised 95 patients with metastatic NSCLC who were treated with ICIs either as the standard of care or in a clinical trial. The following data were extracted from the medical records. The baseline and dynamic neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were calculated in the present study. Responses were assessed by computed tomography (CT) imaging and classified according to the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 every 6-12 weeks during treatment. Results In total, 95 patients were included in the present study. The median age of patients was 61 years, 83.2% (79/95) patients were male, 62.1% (59/95) were former or current smokers, 66.3% (63/95) had adenocarcinoma, 93.7% (89/95) had stage IV disease, and 87.4% were without molecular alterations. A higher overall response rate (ORR) and prolonged median progression-free survival (PFS) was observed in patients with a lower cycle 3 (C3) NLR [7.7 vs. 5.5 months, hazard ratio (HR): 1.70, 95% confidence interval (CI): 0.90-3.22; P=0.12] and derived NLR (dNLR) (8.2 vs. 5.6 months, HR: 1.67, 95% CI: 0.94-2.97; P=0.08). After two cycles of ICI treatment, patients who had an increased NLR, dNLR, and PLR had a lower ORR and an inferior median PFS than those with a decreased NLR (5.5 vs. 8.5 months, HR: 1.87, 95% CI: 1.09-3.21; P=0.02), dNLR (5.6 vs. 8.4 months, HR: 1.49, 95% CI: 0.87-2.57; P=0.15), and PLR (11.8 vs. 5.5 months, HR: 2.28, 95% CI: 1.32-3.94; P=0.003). Moreover, patients with both an increased NLR and PLR had a worse ORR and median PFS than those with either an increased NLR or PLR, or both an increased NLR and PLR (11.8 vs. 5.5 vs. 5.6 months, P=0.003). In addition, the dynamic changes in the PLR could serve as an independent predictive factor of PFS in NSCLC patients treated with ICIs. Conclusions Elevated dynamic changes in the NLR and PLR were associated with lower response rates and shorter PFS in the patients with NSCLC treated with ICIs. Our results also highlight the role of dynamic changes in the PLR in identifying patients with NSCLC who could benefit from ICIs.
Collapse
Affiliation(s)
- Liang Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanjuan Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xinru Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Chenlei Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fei Zhou
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Anwen Xiong
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
10
|
Putzu C, Serra R, Campus R, Fadda GM, Sini C, Marongiu A, Ginesu GC, Fois AG, Palmieri G, Zinellu A, Cossu A, Paliogiannis P. Complete Blood Count-Based Biomarkers as Predictors of Clinical Outcomes in Advanced Non-Small Cell Lung Cancer Patients with PD-L1 < 50% Treated with First-Line Chemoimmunotherapy. Curr Oncol 2024; 31:4955-4967. [PMID: 39329995 PMCID: PMC11431676 DOI: 10.3390/curroncol31090367] [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: 05/22/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Background: The aim of the study was to investigate a series of complete blood cell count-based biomarkers of systemic inflammation as predictors of clinical outcomes in patients who underwent first-line chemoimmunotherapy for advanced NSCLC. Methods: Consecutive patients with pathologically diagnosed stage III/IV NSCLC and PD-L1 < 50% who underwent first-line chemoimmunotherapy were retrospectively enrolled. The clinical outcomes used for biomarker evaluation were Objective Response Rate (ORR) and Overall Survival (OS). Results: Non-responders had significantly higher values of neutrophil to lymphocyte ratio (NLR, median: 5.36; IQR: 2.78-10.82 vs. 3.31; IQR: 2.15-4.12, p = 0.019), neutrophil to monocyte ratio (NMR, median: 14.00; IQR: 8.82-21.20 vs. 9.20; IQR: 7.45-11.20, p = 0.013), and systemic inflammation index (SII, median: 1395; IQR: 929-3334 vs. 945; IQR: 552-1373, p = 0.025), but only NLR and NMR remained independently associated with clinical response in multivariate logistic regression. In the univariate analysis, white blood cells (OR:1.2202; 95% CI: 1.0339-1.4400, p = 0.019), neutrophils (OR:1.2916; 95% CI: 1.0692-1.5604, p = 0.008), NLR (OR:1.3601: 95% CI: 1.0949-1.6896, p = 0.005) and NMR (OR:1.2159; 95% CI: 1.00396-1.4221, p = 0.015) were significantly associated with survival; Cox regression models confirmed that neutrophils, NLR, and MLR were independently associated with survival; NLR, at a cut-off value of 4.0, showed the better AUC (0.749) in predicting OS. Conclusions: Baseline complete blood cell count biomarkers, especially the NLR, can predict clinical outcomes in patients with advanced NSCLC treated with first-line chemoimmunotherapy.
Collapse
Affiliation(s)
- Carlo Putzu
- Medical Oncology Unit, University Hospital of Sassari (AOU SS), Via Enrico De Nicola 39, 07100 Sassari, Italy (G.M.F.)
| | - Riccardo Serra
- Specialty School of Medical Oncology, University of Cagliari, S.S. 554, Km 4500 Bivio per Sestu, 09042 Cagliari, Italy
| | - Rachele Campus
- Specialty School in Pulmonology and Respiratory Diseases, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy
| | - Giovanni Maria Fadda
- Medical Oncology Unit, University Hospital of Sassari (AOU SS), Via Enrico De Nicola 39, 07100 Sassari, Italy (G.M.F.)
| | - Claudio Sini
- Medical Oncology Unit, Giovanni Paolo II Hospital of Olbia, Via Bazzoni Sircana 1, 07026 Olbia, Italy
| | - Andrea Marongiu
- Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy (G.C.G.)
| | - Giorgio Carlo Ginesu
- Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy (G.C.G.)
| | - Alessandro Giuseppe Fois
- Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy (G.C.G.)
| | - Giuseppe Palmieri
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy; (G.P.)
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy; (G.P.)
| | - Antonio Cossu
- Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy (G.C.G.)
| | - Panagiotis Paliogiannis
- Department of Medicine, Surgery and Pharmacology, University of Sassari, Viale San Pietro 43a, 07100 Sassari, Italy (G.C.G.)
| |
Collapse
|
11
|
Tang M, Zhang Z, Wang P, Zhao F, Miao L, Wang Y, Li Y, Li Y, Gao Z. Advancements in precision nanomedicine design targeting the anoikis-platelet interface of circulating tumor cells. Acta Pharm Sin B 2024; 14:3457-3475. [PMID: 39220884 PMCID: PMC11365446 DOI: 10.1016/j.apsb.2024.04.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/10/2024] [Accepted: 03/13/2024] [Indexed: 09/04/2024] Open
Abstract
Tumor metastasis, the apex of cancer progression, poses a formidable challenge in therapeutic endeavors. Circulating tumor cells (CTCs), resilient entities originating from primary tumors or their metastases, significantly contribute to this process by demonstrating remarkable adaptability. They survive shear stress, resist anoikis, evade immune surveillance, and thwart chemotherapy. This comprehensive review aims to elucidate the intricate landscape of CTC formation, metastatic mechanisms, and the myriad factors influencing their behavior. Integral signaling pathways, such as integrin-related signaling, cellular autophagy, epithelial-mesenchymal transition, and interactions with platelets, are examined in detail. Furthermore, we explore the realm of precision nanomedicine design, with a specific emphasis on the anoikis‒platelet interface. This innovative approach strategically targets CTC survival mechanisms, offering promising avenues for combatting metastatic cancer with unprecedented precision and efficacy. The review underscores the indispensable role of the rational design of platelet-based nanomedicine in the pursuit of restraining CTC-driven metastasis.
Collapse
Affiliation(s)
- Manqing Tang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhijie Zhang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuming Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingpeng Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
12
|
Ma J, Pang Y, Shang Y, Xie C, Xu X, Chan L, Zhang Z, Wang W. CyTOF analysis revealed platelet heterogeneity in breast cancer patients received T-DM1 treatment. Clin Immunol 2024; 263:110227. [PMID: 38643891 DOI: 10.1016/j.clim.2024.110227] [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: 01/14/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
T-DM1 (Trastuzumab Emtansine) belongs to class of Antibody-Drug Conjugates (ADC), where cytotoxic drugs are conjugated with the antibody Trastuzumab to specifically target HER2-positive cancer cells. Platelets, as vital components of the blood system, intricately influence the immune response to tumors through complex mechanisms. In our study, we examined platelet surface proteins in the plasma of patients before and after T-DM1 treatment, categorizing them based on treatment response. We identified a subgroup of platelets with elevated expression of CD63 and CD9 exclusively in patients with favorable treatment responses, while this subgroup was absent in patients with poor responses. Another noteworthy discovery was the elevated expression of CD36 in the platelet subgroups of patients exhibiting inadequate responses to treatment. These findings suggest that the expression of these platelet surface proteins may be correlated with the prognosis of T-DM1 treatment. These indicators offer valuable insights for predicting the therapeutic response to T-DM1 and may become important references in future clinical practice, contributing to a better understanding of the impact of ADC therapies and optimizing personalized cancer treatment strategies.
Collapse
Affiliation(s)
- Jianli Ma
- Department of Radiation Oncology, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Yuheng Pang
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Yuefeng Shang
- Department of Breast Surgery, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang, PR China
| | - Chufei Xie
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, PR China
| | - Xiaoxue Xu
- Capital Medical University, Beijing, PR China
| | - Liujia Chan
- Capital Medical University, Beijing, PR China
| | - Zhiren Zhang
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin 150001, China.
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
13
|
Aydın AA, Topçugil F. The Potential Adverse Impact of Post-Treatment Thrombocytopenia on Clinical Outcomes in Cancer Patients Treated With Immune Checkpoint Inhibitors. Cureus 2024; 16:e62163. [PMID: 38993472 PMCID: PMC11238761 DOI: 10.7759/cureus.62163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND The main goal of this study is to explore the prognostic and predictive implications of post-treatment thrombocytopenia on treatment efficacy and clinical outcomes in advanced-stage cancer treated with immune checkpoint inhibitors (ICIs). METHODS This retrospective study included 102 patients with advanced-stage cancer who were treated with ICIs. The simultaneous administration of chemotherapy and ICIs was omitted; nevertheless, the selection of chemotherapy agents employed in different treatment lines was left to the discretion of the attending clinician. Patients were stratified into distinct cohorts based on their post-treatment platelet counts (evaluated for up to four to six months after the completion of ICI). The primary endpoint of interest was progression-free survival (PFS), and overall survival (OS) was the secondary endpoint. RESULTS Patients with superior Eastern Cooperative Oncology Group (ECOG) performance status and those who received ICI as second-line treatment displayed markedly elevated incidences of grade 1 thrombocytopenia (p < 0.05). Kaplan-Meier survival analysis confirmed that patients with high-grade thrombocytopenia had significantly shorter PFS (six vs. 13 vs. 19 months, p < 0.0001) and OS (10 vs. 21 vs. 25 months, p < 0.0001) than those with lower grades or without thrombocytopenia, respectively. Multivariate analysis revealed that decreased platelet levels were a negative independent prognostic factor for both PFS and OS in patients with advanced-stage cancer who received ICIs. CONCLUSION The results of this retrospective study suggest that a decline in platelet levels after treatment represents a dependable adverse prognostic biomarker for clinical outcomes. Moreover, a decrease in platelet levels has been linked to reduced treatment efficacy in advanced-stage cancer patients receiving ICIs, thereby providing valuable prognostic insights for the implementation of personalized treatment strategies in cancer immunotherapy.
Collapse
Affiliation(s)
- Asım Armağan Aydın
- Department of Medical Oncology, Antalya Education and Research Hospital, Antalya, TUR
| | - Füsun Topçugil
- Department of Internal Medicine, Batıgöz Health Group Private Hospital, İzmir, TUR
| |
Collapse
|
14
|
Su Z, Tang J, He Y, Zeng WH, Yu Q, Cao XL, Zou GR. Pan‑immune‑inflammation value as a novel prognostic biomarker in nasopharyngeal carcinoma. Oncol Lett 2024; 27:252. [PMID: 38646495 PMCID: PMC11027095 DOI: 10.3892/ol.2024.14385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 02/06/2024] [Indexed: 04/23/2024] Open
Abstract
The pan-immune-inflammation-value (PIV) is a comprehensive biomarker that integrates different peripheral blood cell subsets. The present study aimed to evaluate the prognostic ability of PIV in patients with nasopharyngeal carcinoma (NPC) undergoing chemoradiotherapy. PIV was assessed using the following equation: (Neutrophil count × platelet count × monocyte count)/lymphocyte count. The Kaplan-Meier method and Cox hazards regression models were used for survival analyses. The optimal cut-off values for PIV and systemic immune-inflammation index (SII) were determined using receiver operating characteristic analysis to be 428.0 and 1032.7, respectively. A total of 319 patients were recruited. Patients with a low baseline PIV (≤428.0) accounted for 69.9% (n=223) and patients with a high baseline PIV (>428.0) accounted for 30.1% (n=96). Compared with patients with low PIV, patients with a high PIV had significantly worse 5-year progression-free survival [PFS; 66.8 vs. 77.1%; hazard ratio (HR), 1.97; 95% confidence interval (CI), 1.22-3.23); P=0.005] and 5-year overall survival (OS; 68.7 vs. 86.9%, HR, 2.71; 95% CI, 1.45-5.03; P=0.001). PIV was also a significant independent prognostic indicator for OS (HR, 2.19; 95% CI, 1.16-4.12; P=0.016) and PFS (HR, 1.86; 95% CI, 1.14-3.04; P=0.013) and outperformed the SII in multivariate analysis. In conclusion, the PIV was a powerful predictor of survival outcomes and outperformed the SII in patients with NPC treated with chemoradiotherapy. Prospective validation of the PIV should be performed to better stratify radical treatment of patients with NPC.
Collapse
Affiliation(s)
- Zhen Su
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| | - Jie Tang
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
- Department of Radiotherapy, Jinan University, Guangzhou, Guangdong 511400, P.R. China
| | - Yan He
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| | - Wei Hua Zeng
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| | - Qian Yu
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| | - Xiao Long Cao
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| | - Guo Rong Zou
- Department of Oncology, Panyu Central Hospital, Cancer Institute of Panyu, Guangzhou, Guangdong 511400, P.R. China
| |
Collapse
|
15
|
Garcia-Leon MJ, Liboni C, Mittelheisser V, Bochler L, Follain G, Mouriaux C, Busnelli I, Larnicol A, Colin F, Peralta M, Osmani N, Gensbittel V, Bourdon C, Samaniego R, Pichot A, Paul N, Molitor A, Carapito R, Jandrot-Perrus M, Lefebvre O, Mangin PH, Goetz JG. Platelets favor the outgrowth of established metastases. Nat Commun 2024; 15:3297. [PMID: 38740748 DOI: 10.1038/s41467-024-47516-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/26/2024] [Indexed: 05/16/2024] Open
Abstract
Despite abundant evidence demonstrating that platelets foster metastasis, anti-platelet agents have low therapeutic potential due to the risk of hemorrhages. In addition, whether platelets can regulate metastasis at the late stages of the disease remains unknown. In this study, we subject syngeneic models of metastasis to various thrombocytopenic regimes to show that platelets provide a biphasic contribution to metastasis. While potent intravascular binding of platelets to tumor cells efficiently promotes metastasis, platelets further support the outgrowth of established metastases via immune suppression. Genetic depletion and pharmacological targeting of the glycoprotein VI (GPVI) platelet-specific receptor in humanized mouse models efficiently reduce the growth of established metastases, independently of active platelet binding to tumor cells in the bloodstream. Our study demonstrates therapeutic efficacy when targeting animals bearing growing metastases. It further identifies GPVI as a molecular target whose inhibition can impair metastasis without inducing collateral hemostatic perturbations.
Collapse
Affiliation(s)
- Maria J Garcia-Leon
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Paris, France.
- Domain therapeutics, Parc d'Innovation - 220 Boulevard Gonthier D'Andernach, 67400, Strasbourg - Illkirch, France.
| | - Cristina Liboni
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Vincent Mittelheisser
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Louis Bochler
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Gautier Follain
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Clarisse Mouriaux
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France
| | - Ignacio Busnelli
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Annabel Larnicol
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Florent Colin
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Marina Peralta
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Naël Osmani
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Valentin Gensbittel
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Catherine Bourdon
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France
| | - Rafael Samaniego
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM), Unidad de Microscopía Confocal, Madrid, Spain
| | - Angélique Pichot
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Nicodème Paul
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Anne Molitor
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
| | - Raphaël Carapito
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Laboratoire d'ImmunoRhumatologie Moléculaire, Plateforme GENOMAX, Institut national de la santé et de la recherche médicale (INSERM) UMR_S 1109, Institut thématique interdisciplinaire (ITI) de Médecine de Précision de Strasbourg Transplantex NG, Faculté de Médecine, France
- Service d'Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, 1 Place de l'Hôpital, 67091, Strasbourg, France
| | | | - Olivier Lefebvre
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
- Equipe Labellisée Ligue Contre le Cancer, Paris, France
| | - Pierre H Mangin
- UMR_S 1255, INSERM, Etablissement Français du Sang-Alsace, Université de Strasbourg, F-67000, Strasbourg, France.
| | - Jacky G Goetz
- Tumor Biomechanics, INSERM UMR_S1109, Strasbourg, France.
- Université de Strasbourg, Strasbourg, France.
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France.
- Equipe Labellisée Ligue Contre le Cancer, Paris, France.
| |
Collapse
|
16
|
Ning Z, Liu K, Zhang H, Dong G, Wang X, Xiong H. Platelets induce CD39 expression in tumor cells to facilitate tumor metastasis. Br J Cancer 2024; 130:1542-1551. [PMID: 38461171 PMCID: PMC11058827 DOI: 10.1038/s41416-024-02640-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 03/11/2024] Open
Abstract
BACKGROUND Tumor cells continue to evolve the metastatic potential in response to signals provided by the external microenvironment during metastasis. Platelets closely interact with tumor cells during hematogenous metastasis and facilitate tumor development. However, the molecular mechanisms underlying this process are not fully understood. METHODS RNA-sequencing was performed to screen differentially expressed genes mediated by platelets. The effects of platelet and CD39 on tumor metastasis were determined by experimental metastasis models with WT, NCG and CD39-/- mice. RESULTS RNA-sequencing results showed that platelets significantly up-regulated CD39 expression in tumor cells. CD39 is a novel immune checkpoint molecule and a key driver of immunosuppression. Our data provided evidence that the expression of CD39 was enhanced by platelets in a platelet-tumor cell contact dependent manner. Although the role of CD39 expressed by immune cells is well established, the effect of CD39 expressed by tumor cells on tumor cell behavior, anti-tumor immunity and tumor metastasis is unclear. We found that CD39 promoted tumor cell invasion, but had no effect on proliferation and migration. Notably, we showed that the ability of platelets to prime tumor cells for metastasis depends on CD39 in the experimental tumor metastasis model. CD39 silencing resulted in fewer experimental metastasis formation, and this anti-metastasis effect was significantly reduced in platelet-depleted mice. Furthermore, overexpression of CD39 in tumor cells promoted metastasis. In order to eliminate the effect of CD39 expressed in cells other than tumor cells, we detected tumor metastasis in CD39-/- mice and obtained similar results. Moreover, overexpression of CD39 in tumor cells inhibited antitumor immunity. Finally, the data from human samples also supported our findings. CONCLUSIONS Our study shows that direct contact with platelets induces CD39 expression in tumor cells, leading to immune suppression and promotion of metastasis.
Collapse
Affiliation(s)
- Zhaochen Ning
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Keyan Liu
- Department of Public Health, Jining Medical University, Jining, 272067, China
| | - Hui Zhang
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Guanjun Dong
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Xiaotong Wang
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China
| | - Huabao Xiong
- Jining Key Laboratory of Immunology, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
17
|
Chen SJ, Hashimoto K, Fujio K, Hayashi K, Paul SK, Yuzuriha A, Qiu WY, Nakamura E, Kanashiro MA, Kabata M, Nakamura S, Sugimoto N, Kaneda A, Yamamoto T, Saito H, Takayama N, Eto K. A let-7 microRNA-RALB axis links the immune properties of iPSC-derived megakaryocytes with platelet producibility. Nat Commun 2024; 15:2588. [PMID: 38519457 PMCID: PMC10960040 DOI: 10.1038/s41467-024-46605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/04/2024] [Indexed: 03/25/2024] Open
Abstract
We recently achieved the first-in-human transfusion of induced pluripotent stem cell-derived platelets (iPSC-PLTs) as an alternative to standard transfusions, which are dependent on donors and therefore variable in supply. However, heterogeneity characterized by thrombopoiesis-biased or immune-biased megakaryocytes (MKs) continues to pose a bottleneck against the standardization of iPSC-PLT manufacturing. To address this problem, here we employ microRNA (miRNA) switch biotechnology to distinguish subpopulations of imMKCLs, the MK cell lines producing iPSC-PLTs. Upon miRNA switch-based screening, we find imMKCLs with lower let-7 activity exhibit an immune-skewed transcriptional signature. Notably, the low activity of let-7a-5p results in the upregulation of RAS like proto-oncogene B (RALB) expression, which is crucial for the lineage determination of immune-biased imMKCL subpopulations and leads to the activation of interferon-dependent signaling. The dysregulation of immune properties/subpopulations, along with the secretion of inflammatory cytokines, contributes to a decline in the quality of the whole imMKCL population.
Collapse
Affiliation(s)
- Si Jing Chen
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazuya Hashimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Kosuke Fujio
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Karin Hayashi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sudip Kumar Paul
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akinori Yuzuriha
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Wei-Yin Qiu
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Emiri Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | | | - Mio Kabata
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Sou Nakamura
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Naoshi Sugimoto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takuya Yamamoto
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Medical-risk Avoidance Based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto, Japan
| | - Hirohide Saito
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Koji Eto
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
- Department of Regenerative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.
| |
Collapse
|
18
|
Tassinari S, D'Angelo E, Caicci F, Grange C, Burrello J, Fassan M, Brossa A, Bao RQ, Spolverato G, Agostini M, Collino F, Bussolati B. Profile of matrix-entrapped extracellular vesicles of microenvironmental and infiltrating cell origin in decellularized colorectal cancer and adjacent mucosa. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e144. [PMID: 38939413 PMCID: PMC11080771 DOI: 10.1002/jex2.144] [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: 08/01/2023] [Revised: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 06/29/2024]
Abstract
Cellular elements that infiltrate and surround tumours and pre-metastatic tissues have a prominent role in tumour invasion and growth. The extracellular vesicles specifically entrapped and stored within the extracellular matrix (ECM-EVs) may reflect the different populations of the tumour microenvironment and their change during tumour progression. However, their profile is at present unknown. To elucidate this aspect, we isolated and characterized EVs from decellularized surgical specimens of colorectal cancer and adjacent colon mucosa and analyzed their surface marker profile. ECM-EVs in tumours and surrounding mucosa mainly expressed markers of lymphocytes, natural killer cells, antigen-presenting cells, and platelets, as well as epithelial cells, representing a multicellular microenvironment. No difference in surface marker expression was observed between tumour and mucosa ECM-EVs in stage II-III tumours. At variance, in the colon mucosa adjacent to stage IV carcinomas, ECM-EV profile showed a significantly increased level of immune, epithelial and platelet markers in comparison to the matrix of the corresponding tumour. The increase of EVs from immune cells and platelets was not observed in the mucosa adjacent to low-stage tumours. In addition, CD25, a T-lymphocyte marker, resulted specifically overexpressed by ECM-EVs from stage IV carcinomas, possibly correlated with the pro-tolerogenic environment found in the corresponding tumour tissue. These results outline the tissue microenvironmental profile of EVs in colorectal carcinoma-derived ECM and unveil a profound change in the healthy mucosa adjacent to high-stage tumours.
Collapse
Affiliation(s)
- Sarah Tassinari
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| | - Edoardo D'Angelo
- General Surgery 3, Department of SurgeryOncology and Gastroenterology, University of PadovaPaduaItaly
- NanoInspired biomedicine lab, Fondazione Istituto di Ricerca Pediatrica Città della SperanzaPaduaItaly
| | | | - Cristina Grange
- Department of Medical ScienceUniversity of TorinoTorinoItaly
| | - Jacopo Burrello
- Department of Medical ScienceUniversity of TorinoTorinoItaly
| | - Matteo Fassan
- Department of Medicine (DIMED)University of PaduaPaduaItaly
- Veneto Institute of Oncology IOV ‐ IRCCSPaduaItaly
| | - Alessia Brossa
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| | - Riccardo Quoc Bao
- General Surgery 3, Department of SurgeryOncology and Gastroenterology, University of PadovaPaduaItaly
| | - Gaya Spolverato
- General Surgery 3, Department of SurgeryOncology and Gastroenterology, University of PadovaPaduaItaly
| | - Marco Agostini
- General Surgery 3, Department of SurgeryOncology and Gastroenterology, University of PadovaPaduaItaly
- NanoInspired biomedicine lab, Fondazione Istituto di Ricerca Pediatrica Città della SperanzaPaduaItaly
| | - Federica Collino
- Laboratory of Translational Research in Paediatric Nephro‐UrologyFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanoItaly
- Pediatric Nephrology, Dialysis and Transplant UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanoItaly
- Department of Clinical Sciences and Community HealthUniversity of MilanoMilanItaly
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| |
Collapse
|
19
|
Mierke CT. Phenotypic Heterogeneity, Bidirectionality, Universal Cues, Plasticity, Mechanics, and the Tumor Microenvironment Drive Cancer Metastasis. Biomolecules 2024; 14:184. [PMID: 38397421 PMCID: PMC10887446 DOI: 10.3390/biom14020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor diseases become a huge problem when they embark on a path that advances to malignancy, such as the process of metastasis. Cancer metastasis has been thoroughly investigated from a biological perspective in the past, whereas it has still been less explored from a physical perspective. Until now, the intraluminal pathway of cancer metastasis has received the most attention, while the interaction of cancer cells with macrophages has received little attention. Apart from the biochemical characteristics, tumor treatments also rely on the tumor microenvironment, which is recognized to be immunosuppressive and, as has recently been found, mechanically stimulates cancer cells and thus alters their functions. The review article highlights the interaction of cancer cells with other cells in the vascular metastatic route and discusses the impact of this intercellular interplay on the mechanical characteristics and subsequently on the functionality of cancer cells. For instance, macrophages can guide cancer cells on their intravascular route of cancer metastasis, whereby they can help to circumvent the adverse conditions within blood or lymphatic vessels. Macrophages induce microchannel tunneling that can possibly avoid mechanical forces during extra- and intravasation and reduce the forces within the vascular lumen due to vascular flow. The review article highlights the vascular route of cancer metastasis and discusses the key players in this traditional route. Moreover, the effects of flows during the process of metastasis are presented, and the effects of the microenvironment, such as mechanical influences, are characterized. Finally, the increased knowledge of cancer metastasis opens up new perspectives for cancer treatment.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth System Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
20
|
Zhang C, Qin C, Dewanjee S, Bhattacharya H, Chakraborty P, Jha NK, Gangopadhyay M, Jha SK, Liu Q. Tumor-derived small extracellular vesicles in cancer invasion and metastasis: molecular mechanisms, and clinical significance. Mol Cancer 2024; 23:18. [PMID: 38243280 PMCID: PMC10797874 DOI: 10.1186/s12943-024-01932-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/02/2024] [Indexed: 01/21/2024] Open
Abstract
The production and release of tumor-derived small extracellular vesicles (TDSEVs) from cancerous cells play a pivotal role in the propagation of cancer, through genetic and biological communication with healthy cells. TDSEVs are known to orchestrate the invasion-metastasis cascade via diverse pathways. Regulation of early metastasis processes, pre-metastatic niche formation, immune system regulation, angiogenesis initiation, extracellular matrix (ECM) remodeling, immune modulation, and epithelial-mesenchymal transition (EMT) are among the pathways regulated by TDSEVs. MicroRNAs (miRs) carried within TDSEVs play a pivotal role as a double-edged sword and can either promote metastasis or inhibit cancer progression. TDSEVs can serve as excellent markers for early detection of tumors, and tumor metastases. From a therapeutic point of view, the risk of cancer metastasis may be reduced by limiting the production of TDSEVs from tumor cells. On the other hand, TDSEVs represent a promising approach for in vivo delivery of therapeutic cargo to tumor cells. The present review article discusses the recent developments and the current views of TDSEVs in the field of cancer research and clinical applications.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China
| | - Chaoying Qin
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Niraj Kumar Jha
- Centre of Research Impact and Outreach, Chitkara University Institute of Engineering and Technology, Chitkara University, Punjab, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
| | - Moumita Gangopadhyay
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat, Kolkata, 700126, West Bengal, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, New Delhi, Delhi, 110008, India.
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
- The Institute of Skull Base Surgery and Neuro-Oncology at Hunan Province, Changsha, 410008, China.
| |
Collapse
|
21
|
Katolkar UN, Surana SJ. Exploring the Potential Role of Phytopharmaceuticals in Alleviating Toxicities of Chemotherapeutic Agents. Curr Protein Pept Sci 2024; 25:753-779. [PMID: 38919003 DOI: 10.2174/0113892037307940240606075208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Chemotherapy is the mainstay of cancer treatment, bringing patients optimism about recurrence and survival. However, the clinical effectiveness of chemotherapeutic drugs is frequently jeopardized by their intrinsic toxicity, resulting in side effects affecting the quality of life of cancer patients. This analysis explores the ethnopharmacological impact of phytopharmaceuticals, highlighting their traditional use in many cultures. The present study, which takes its cues from indigenous knowledge, aims to close the knowledge gap between traditional medicine and modern medicine in reducing the toxicities of chemotherapy treatments. AIM The present in-depth study aims to highlight the current research and upcoming developments in phytopharmaceuticals for reducing the toxicity of chemotherapeutic drugs. Further, we address the mechanisms through which phytopharmaceuticals may reduce chemotherapy-induced side effects that include nausea, vomiting, myelosuppression, nephropathy, neuropathy, and cardiotoxicity using data from a variety of preclinical and clinical investigations. MATERIALS AND METHODS The literature search was carried out by employing search engines such as PubMed and Google Scholar with keywords such as cancer, chemotherapy, CNS toxicity, hematopoietic toxicity, renal toxicity, GI toxicity, CNS toxicity, and phytopharmaceuticals. RESULTS Bioactive chemicals found in plants, such as fruits, vegetables, herbs, and spices, are being studied for their capacity to improve the safety and acceptability of chemotherapy regimens. The current review also dives into the investigation of phytopharmaceuticals as adjuvant medicines in cancer treatment, which is a viable path for addressing the pressing need to lessen chemotherapy-induced toxicities. CONCLUSION The present review revealed that the potential of phytopharmaceuticals in alleviating chemotherapeutic drug toxicities would pave the way for better cancer treatment and patient outcomes, harmonizing with the larger trend towards personalized and holistic approaches to chemotherapy.
Collapse
Affiliation(s)
- Ujwal N Katolkar
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| | - Sanjay J Surana
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| |
Collapse
|
22
|
He D, Du S, He S, Song H, Pu B, Zhang G, Yang C. Effect of dynamic platelet-to-lymphocyte ratio on the prognosis of patients with esophageal squamous cell carcinoma receiving chemoradiotherapy. Medicine (Baltimore) 2023; 102:e36554. [PMID: 38065887 PMCID: PMC10713128 DOI: 10.1097/md.0000000000036554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023] Open
Abstract
Systemic inflammatory load affects the long-term developmental outcomes in patients with malignancy. The purpose of this study was to investigate the effect of the dynamic levels of platelet-to-lymphocyte ratio (PLR) at different treatment stages on the prognosis of patients with esophageal squamous cell carcinoma (ESCC) undergoing chemoradiotherapy. This study included 168 patients who received chemoradiotherapy between 2012 and 2018. PLR levels at different treatment stages were calculated based on blood test results. The association between PLR and overall survival (OS) was determined using the Kaplan-Meier method and Cox proportional regression models. The cutoff values of PLR before and after treatment of 168 patients with ESCC were 195.7 and 403.6, respectively. The 5-year OS rates of patients in the low and high pre-PLR groups were 42.1% and 21.7%, respectively. The overall 5-year OS rate of all patients was 27.1%. Multivariate analysis results showed that patient age (hazard ratio [HR] = 1.736; 95% confidence interval (CI) = 1.129-2.669; P = .012), alcohol consumption (HR = 1.622; 95%CI = 1.050-2.508; P = .029), T stage (HR = 12.483; 95%CI = 3.719-41.896; P < .001), pre-PLR (HR = 1.716; 95%CI = 1.069-2.756; P = .025), post-PLR (HR = 1.664; 95%CI = 1.106-2.503; P = .015) were independent factors of the prognosis of patients with ESCC. PLR at different treatment stages can be used to effectively evaluate the prognosis of patients with ESCC undergoing chemoradiotherapy.
Collapse
Affiliation(s)
- Dan He
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| | - Shulan Du
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| | - Songyuan He
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| | - Hao Song
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| | - Bo Pu
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| | - Guojun Zhang
- Nanchong Central Hospital, Gaoping District, Nanchong City, Sichuan Province, China
| | - Chuan Yang
- Guangyuan Central Hospital, Lizhou District, Guangyuan City, China
| |
Collapse
|
23
|
Zhao J, Huang A, Zeller J, Peter K, McFadyen JD. Decoding the role of platelets in tumour metastasis: enigmatic accomplices and intricate targets for anticancer treatments. Front Immunol 2023; 14:1256129. [PMID: 38106409 PMCID: PMC10722285 DOI: 10.3389/fimmu.2023.1256129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
The canonical role of platelets as central players in cardiovascular disease by way of their fundamental role in mediating thrombosis and haemostasis is well appreciated. However, there is now a large body of experimental evidence demonstrating that platelets are also pivotal in various physiological and pathophysiological processes other than maintaining haemostasis. Foremost amongst these is the emerging data highlighting the key role of platelets in driving cancer growth, metastasis and modulating the tumour microenvironment. As such, there is significant interest in targeting platelets therapeutically for the treatment of cancer. Therefore, the purpose of this review is to provide an overview of how platelets contribute to the cancer landscape and why platelets present as valuable targets for the development of novel cancer diagnosis tools and therapeutics.
Collapse
Affiliation(s)
- Jessie Zhao
- Department of Clinical Haematology, Alfred Hospital, Melbourne, VI, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VI, Australia
| | - Angela Huang
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VI, Australia
| | - Johannes Zeller
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VI, Australia
- Department of Plastic and Hand Surgery, Medical Center – University of Freiburg, Medical Faculty of the University of Freiburg, Freiburg, Germany
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VI, Australia
- Department of Cardiology, Alfred Hospital, Melbourne, VI, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Parkville, VI, Australia
- Department of Medicine, Monash University, Melbourne, VI, Australia
| | - James D. McFadyen
- Department of Clinical Haematology, Alfred Hospital, Melbourne, VI, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, VI, Australia
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VI, Australia
- Department of Cardiometabolic Health, The University of Melbourne, Parkville, VI, Australia
| |
Collapse
|
24
|
Wu Y, Zhao J, Wang Z, Liu D, Tian C, Ye B, Sun Y, Li H, Wang X. Association of systemic inflammatory markers and tertiary lymphoid structure with pathological complete response in gastric cancer patients receiving preoperative treatment: a retrospective cohort study. Int J Surg 2023; 109:4151-4161. [PMID: 38259000 PMCID: PMC10720847 DOI: 10.1097/js9.0000000000000741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/24/2023] [Indexed: 01/24/2024]
Abstract
BACKGROUND Assessment of systemic and local immune responses is crucial in determining the efficacy of cancer interventions. The identification of specific factors that correlate with pathological complete response (pCR) is essential for optimizing treatment decisions. METHODS In this retrospective study, a total of 521 patients diagnosed with gastric adenocarcinoma who underwent curative gastrectomy following preoperative treatment were reviewed. Of these patients, 463 did not achieve pCR (non-pCR) and 58 achieved pCR. Clinicopathological factors were evaluated to identify predictors for pCR using a logistic regression model. Additionally, a smaller cohort (n=76) was derived using propensity score matching to investigate local immune response, specifically the features of tertiary lymphoid structure (TLS) using H&E staining, immunohistochemistry, and multiplex immunofluorescence. RESULTS The multivariate regression analysis demonstrated a significant association between low systemic inflammatory status and pCR, as evidenced by reduced levels of the combined systemic immune-inflammation index (SII) and neutrophil-to-lymphocyte ratio (NLR) (SII+NLR) (odds ratio: 3.33, 95% CI: 1.79-6.17, P<0.001). In the smaller cohort analysis, distinct TLS characteristics were correlated with the presence of pCR. Specifically, a higher density of TLS and a lower proportion of PD1+ cells and CD8+ cells within TLS in the tumor bed were strongly associated with pCR. CONCLUSION Both systemic and local immune profile were associated with pCR. A low level of SII+NLR served as an independent predictor of pCR, while distinct TLS features were associated with the presence of pCR. Focusing on the immune profile was crucial for optimal management of gastric cancer patients receiving preoperative treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Haojie Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
25
|
Guha A, Goswami KK, Sultana J, Ganguly N, Choudhury PR, Chakravarti M, Bhuniya A, Sarkar A, Bera S, Dhar S, Das J, Das T, Baral R, Bose A, Banerjee S. Cancer stem cell-immune cell crosstalk in breast tumor microenvironment: a determinant of therapeutic facet. Front Immunol 2023; 14:1245421. [PMID: 38090567 PMCID: PMC10711058 DOI: 10.3389/fimmu.2023.1245421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer (BC) is globally one of the leading killers among women. Within a breast tumor, a minor population of transformed cells accountable for drug resistance, survival, and metastasis is known as breast cancer stem cells (BCSCs). Several experimental lines of evidence have indicated that BCSCs influence the functionality of immune cells. They evade immune surveillance by altering the characteristics of immune cells and modulate the tumor landscape to an immune-suppressive type. They are proficient in switching from a quiescent phase (slowly cycling) to an actively proliferating phenotype with a high degree of plasticity. This review confers the relevance and impact of crosstalk between immune cells and BCSCs as a fate determinant for BC prognosis. It also focuses on current strategies for targeting these aberrant BCSCs that could open avenues for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Aishwarya Guha
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | | | - Jasmine Sultana
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nilanjan Ganguly
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Pritha Roy Choudhury
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Mohona Chakravarti
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Avishek Bhuniya
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anirban Sarkar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Saurav Bera
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sukanya Dhar
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Juhina Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Tapasi Das
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Rathindranath Baral
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| | - Anamika Bose
- Department of Pharmaceutical Technology Biotechnology National Institute of Pharmaceutical Education and Research (NIPER) Sahibzada Ajit Singh (S.A.S.) Nagar, Mohali, Punjab, India
| | - Saptak Banerjee
- Department of Immunoregulation and Immunodiagnostics, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
26
|
Yang L, Zhang K, Zheng D, Bai Y, Yue D, Wu L, Ling H, Ni S, Zou H, Ye B, Liu C, Deng Y, Liu Q, Li Y, Wang D. Platelet-Based Nanoparticles with Stimuli-Responsive for Anti-Tumor Therapy. Int J Nanomedicine 2023; 18:6293-6309. [PMID: 37954456 PMCID: PMC10637234 DOI: 10.2147/ijn.s436373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
In addition to hemostasis and coagulation, years of studies have proved that platelets are involved in the whole process of tumor progression, including tumor invasion, intravasation, extravasation, and so on. It means that this property of platelets can be used in anti-tumor therapy. However, traditional platelet-based antitumor drugs often cause autologous platelet damage due to lack of targeting, resulting in serious side effects. Therefore, the researchers designed a variety of anti-tumor drug delivery systems based on platelets by targeting platelets or platelet membrane coating. The drug delivery systems have special response modes, which is crucial in the design of nanoparticles. These modes enhance the targeting and improve the anti-tumor effect. Here, we present a review of recent discoveries in the field of the crosstalk between platelets and tumors and the progress of platelet-based anti-tumor nanoparticles.
Collapse
Affiliation(s)
- Linlan Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Kaijiong Zhang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Dongming Zheng
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Yuxin Bai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Daifan Yue
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Lichun Wu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Han Ling
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Sujiao Ni
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Haimin Zou
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Bo Ye
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Chang Liu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yao Deng
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Qiancheng Liu
- Department of Clinical Laboratory of Mianyang People’s Hospital, Mianyang, People’s Republic of China
| | - Yan Li
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
| | - Dongsheng Wang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, People’s Republic of China
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
27
|
Kou J, Huang J, Li J, Wu Z, Ni L. Systemic immune-inflammation index predicts prognosis and responsiveness to immunotherapy in cancer patients: a systematic review and meta‑analysis. Clin Exp Med 2023; 23:3895-3905. [PMID: 36966477 DOI: 10.1007/s10238-023-01035-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 03/01/2023] [Indexed: 03/27/2023]
Abstract
The systemic immune-inflammation index (SII) is a significant prognostic factor in some cancer types. However, the prognostic role of SII in cancer patients with immunotherapy remains uncertain. We aimed to evaluate the relationship between pretreatment SII and clinical survival outcomes for advanced-stage cancer patients treated with immune checkpoint inhibitors (ICIs). A comprehensive literature search was performed to identify eligible studies concerning the association between pretreatment SII and survival outcomes in advanced cancer patients treated with ICIs. The data were extracted from publications and used to calculate the pooled odds ratio (pOR) for objective response rate (ORR), disease control rate (DCR), and pooled hazard ratio (pHR) for overall survival (OS), progressive-free survival (PFS), along with 95% confidence intervals (95% CIs). Fifteen articles with 2438 participants were included. A higher level of SII indicated a lower ORR (pOR = 0.73, 95% CI 0.56-0.94) and worse DCR (pOR = 0.56, 95% CI 0.35-0.88). High SII was associated with a shorter OS (pHR = 2.33, 95% CI 2.02-2.69) and unfavorable PFS (pHR = 1.85, 95% CI 1.61-2.14). Therefore, high SII level might be a non-invasive and efficacious biomarker of poor tumor response and adverse prognosis of advanced cancer patients with immunotherapy.
Collapse
Affiliation(s)
- Junyan Kou
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jing Huang
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jun Li
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Zhen Wu
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Liwei Ni
- Department of Oncology, Hangzhou Cancer Hospital, Hangzhou, 310000, Zhejiang, People's Republic of China.
| |
Collapse
|
28
|
Wang C, Li P. Risk factors for intraoperative blood loss in resection of intracranial meningioma: Analysis of 530 cases. PLoS One 2023; 18:e0291171. [PMID: 37682850 PMCID: PMC10490957 DOI: 10.1371/journal.pone.0291171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
PURPOSE Excision of intracranial meningioma has been associated with major intraoperative blood loss (IBL). The objective of the study was to identify factors affecting IBL during removal of meningioma. METHODS We retrospectively studied medical records of 530 adult patients who underwent surgery for intracranial meningioma at Sichuan Provincial People's Hospital between September 2018 and May 2022. We obtained the following data from each patient's medical chart: age, sex, height, weight, comorbidities, blood pressure, history of smoking and alcohol, imaging examination findings, pathologic diagnosis, albumin, creatinine, calcium, magnesium, hemoglobin (Hb), hematocrit, platelet count, activated partial thromboplastin time, international normalized ratio, fibrinogen concentration and blood transfusion. Univariate and multivariate analyses were performed to identify risk factors for greater IBL during removal of intracranial meningioma. RESULTS A total of 530 patients were included in our study. Univariate analysis revealed that sex (p = 0.004), two-dimensional (2D) tumor area (p < 0.001), sinus involvement (p = 0.014), World Health Organization grade (p = 0.015), preoperative albumin level (p = 0.032), preoperative Hb level (p = 0.001) and preoperative platelet count (p = 0.004) were significantly associated with greater IBL. Multivariate analysis revealed that greater 2D tumor area (p < 0.001), higher preoperative albumin concentration (p = 0.029) and higher preoperative platelet count (p = 0.03) were independent risk factors for greater IBL in resection of intracranial meningioma. CONCLUSION Larger tumor size, higher preoperative albumin concentration and higher preoperative platelet count were identified as independent risk factors for greater IBL in resection of intracranial meningioma.
Collapse
Affiliation(s)
- Chenghong Wang
- Department of Anesthesiology, Sichuan Provincial People’ Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Peng Li
- Department of Anesthesiology, Sichuan Provincial People’ Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
29
|
Luo L, Zhang B, Tao F, Chen Z, Ye Q, Zhao X, Wu J. Perfluorotributylamine-Loaded Albumin Nanoparticles Downregulate Platelet-Derived TGFβ to Inhibit Tumor Metastasis. ACS NANO 2023; 17:15388-15400. [PMID: 37526429 DOI: 10.1021/acsnano.3c00295] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
Tumor metastasis contributes to the low overall survival of tumor patients, while transforming growth factor-β (TGFβ) has been recognized as a prominently promoting factor in the development of tumor metastasis. Platelets reserve abundant TGFβ, which will be secreted to peripheral blood after activation, and they are the dominant source of circulating TGFβ. Therefore, downregulation of platelet-derived TGFβ is expected to inhibit the metastasis of circulating tumor cells. Here, unfolded human serum albumin (HSA)-coated perfluorotributylamine (PFTBA) nanoparticles were constructed to display a favorable platelet delivery and an antiplatelet effect to downregulate platelet-derived TGFβ in vitro and in blood plasma. PFTBA@HSA-mediated TGFβ downregulation impaired epithelial-mesenchymal transition of tumor cells as well as their migration and invasion behaviors and enhanced immune surveillance of NK cells. Intravenous injection of PFTBA@HSA effectively reduced tumor metastasis on the lungs or liver to improve the survival rate of mice on multiple metastatic models, including CT26 colon cancer, B16F10 melanoma, and 4T1 breast cancer. Compared with the clinical antiplatelet drug ticagrelor, PFTBA@HSA reduced bleeding risk when displaying a favorable downregulation on platelet-derived TGFβ, thereby obtaining a higher therapy benefit. Together, this study confirmed that downregulation of platelet-derived TGFβ by PFTBA@HSA will be a potential approach and therapeutic candidate for the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Lifeng Luo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
- Department of Urology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210093, China
| | - Baoli Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
| | - Feng Tao
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
| | - Zhong Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
| | - Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
| | - Xiaozhi Zhao
- Department of Urology, Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210093, China
- Wuxi Xishan NJU Institute of Applied Biotechnology, Nanjing University, Wuxi 214101, China
| |
Collapse
|
30
|
Wilhelm G, Mertowska P, Mertowski S, Przysucha A, Strużyna J, Grywalska E, Torres K. The Crossroads of the Coagulation System and the Immune System: Interactions and Connections. Int J Mol Sci 2023; 24:12563. [PMID: 37628744 PMCID: PMC10454528 DOI: 10.3390/ijms241612563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The coagulation and immune systems, two vital systems in the human body, share intimate connections that fundamentally determine patient health. These systems work together through several common regulatory pathways, including the Tissue Factor (TF) Pathway. Immune cells expressing TF and producing pro-inflammatory cytokines can influence coagulation, while coagulation factors and processes reciprocally impact immune responses by activating immune cells and controlling their functions. These shared pathways contribute to maintaining health and are also involved in various pathological conditions. Dysregulated coagulation, triggered by infection, inflammation, or tissue damage, can result in conditions such as disseminated intravascular coagulation (DIC). Concurrently, immune dysregulation may lead to coagulation disorders and thrombotic complications. This review elucidates these intricate interactions, emphasizing their roles in the pathogenesis of autoimmune diseases and cancer. Understanding the complex interplay between these systems is critical for disease management and the development of effective treatments. By exploring these common regulatory mechanisms, we can uncover innovative therapeutic strategies targeting these intricate disorders. Thus, this paper presents a comprehensive overview of the mutual interaction between the coagulation and immune systems, highlighting its significance in health maintenance and disease pathology.
Collapse
Affiliation(s)
- Grzegorz Wilhelm
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Anna Przysucha
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Jerzy Strużyna
- East Center of Burns Treatment and Reconstructive Surgery, Medical University of Lublin, 20-059 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Kamil Torres
- Department of Plastic and Reconstructive Surgery and Microsurgery, Medical University of Lublin, 20-059 Lublin, Poland; (G.W.); (K.T.)
| |
Collapse
|
31
|
Eslami-S Z, Cortés-Hernández LE, Glogovitis I, Antunes-Ferreira M, D’Ambrosi S, Kurma K, Garima F, Cayrefourcq L, Best MG, Koppers-Lalic D, Wurdinger T, Alix-Panabières C. In vitro cross-talk between metastasis-competent circulating tumor cells and platelets in colon cancer: a malicious association during the harsh journey in the blood. Front Cell Dev Biol 2023; 11:1209846. [PMID: 37601099 PMCID: PMC10433913 DOI: 10.3389/fcell.2023.1209846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Background: Platelets are active players in hemostasis, coagulation and also tumorigenesis. The cross-talk between platelets and circulating tumor cells (CTCs) may have various pro-cancer effects, including promoting tumor growth, epithelial-mesenchymal transition (EMT), metastatic cell survival, adhesion, arrest and also pre-metastatic niche and metastasis formation. Interaction with CTCs might alter the platelet transcriptome. However, as CTCs are rare events, the cross-talk between CTCs and platelets is poorly understood. Here, we used our established colon CTC lines to investigate the colon CTC-platelet cross-talk in vitro and its impact on the behavior/phenotype of both cell types. Methods: We exposed platelets isolated from healthy donors to thrombin (positive control) or to conditioned medium from three CTC lines from one patient with colon cancer and then we monitored the morphological and protein expression changes by microscopy and flow cytometry. We then analyzed the transcriptome by RNA-sequencing of platelets indirectly (presence of a Transwell insert) co-cultured with the three CTC lines. We also quantified by reverse transcription-quantitative PCR the expression of genes related to EMT and cancer development in CTCs after direct co-culture (no Transwell insert) with platelets. Results: We observed morphological and transcriptomic changes in platelets upon exposure to CTC conditioned medium and indirect co-culture (secretome). Moreover, the expression levels of genes involved in EMT (p < 0.05) were decreased in CTCs co-cultured with platelets, but not of genes encoding mesenchymal markers (FN1 and SNAI2). The expression levels of genes involved in cancer invasiveness (MYC, VEGFB, IL33, PTGS2, and PTGER2) were increased. Conclusion: For the first time, we studied the CTC-platelet cross-talk using our unique colon CTC lines. Incubation with CTC conditioned medium led to platelet aggregation and activation, supporting the hypothesis that their interaction may contribute to preserve CTC integrity during their journey in the bloodstream. Moreover, co-culture with platelets influenced the expression of several genes involved in invasiveness and EMT maintenance in CTCs.
Collapse
Affiliation(s)
- Zahra Eslami-S
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Luis Enrique Cortés-Hernández
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Ilias Glogovitis
- Department of Neurosurgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Brain Tumor Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Mafalda Antunes-Ferreira
- Department of Neurosurgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Brain Tumor Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Silvia D’Ambrosi
- Department of Neurosurgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Brain Tumor Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Keerthi Kurma
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Françoise Garima
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Myron G. Best
- Department of Neurosurgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Brain Tumor Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | | | - Thomas Wurdinger
- Department of Neurosurgery, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Brain Tumor Center Amsterdam, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells—University Medical Center of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES), Université de Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
32
|
Zhou J, Chu X, Zhao J, Xie M, Wu J, Yu X, Fang Y, Li Y, Li X, Su C. Full spectrum flow cytometry-powered comprehensive analysis of PBMC as biomarkers for immunotherapy in NSCLC with EGFR-TKI resistance. Biol Proced Online 2023; 25:21. [PMID: 37488517 PMCID: PMC10364374 DOI: 10.1186/s12575-023-00215-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/14/2023] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Clinical studies suggest that immune checkpoint inhibitor (ICI) monotherapy has limited benefits in non-small cell lung cancer (NSCLC) patients after epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) failure. However, data about efficacy of ICI plus chemotherapy remain controversial, probably attributed to the heterogeneity among such population, and robust efficacy biomarkers are urgent to explore. METHODS A total of 60 eligible patients who received ICI plus chemotherapy after EGFR-TKI treatment failure were enrolled, 24 of whom peripheral blood mononuclear cell (PBMC) samples were collected at baseline and after 2 cycles of treatment. We have designed a 23-color-antibody panel to detect PBMC by full spectrum flow cytometry. RESULTS For EGFR-TKI resistant NSCLC patients: 1) ICI plus chemotherapy achieved an objective response rate (ORR) of 21.7% and a median progression-free survival (PFS) of 6.4 months. 2) clinical characteristics associated with worse efficacy included liver metastasis and platelet-to-lymphocyte ratio (PLR) > 200. 3) the proportion of immune cell subset associated with better efficacy was higher baseline effective CD4+T cells (E4). 4) the baseline expression of immune checkpoint proteins (ICPs) on cell subsets associated with better efficacy included: higher expression of CD25 on dendritic cells (DC) and central memory CD8+T cells (CM8), and higher expression of Lymphocyte activation gene 3 (LAG-3) on effective memory CD8+T cells (EM8). 5) the expression of ICPs after 2 cycles of treatment associated with better efficacy included: higher expression of CD25 on CD8+T/EM8 /natural killer (NK) cells. 6) the dynamic changes of ICPs expression associated with worse efficacy included: significantly decrease of T cell immunoglobulin and ITIM domain (TIGIT) expression on regular T cells (Tregs) and decrease of V-domain immunoglobulin suppressor of T cell activation (VISTA) expression on Th1. 7) a prediction model for the efficacy of ICI plus chemotherapy was successfully constructed with a sensitivity of 62.5%, specificity of 100%, and area under curve (AUC) = 0.817. CONCLUSIONS Some EGFR-TKI-resistant NSCLC patients could indeed benefit from ICI plus chemotherapy, but most patients are primary resistant to immunotherapy. Comprehensive analysis of peripheral immune cells using full spectrum flow cytometry showed that compared to the proportion of cell subsets, the expression type and level of ICPs on immune cells, especially CD25, were significantly correlated with the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Xiangling Chu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Jing Zhao
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Mengqing Xie
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Jing Wu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Xin Yu
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Yujia Fang
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China
| | - Yazhou Li
- Righton Biotechnology Co., Ltd, Shanghai, China
| | - Xiyan Li
- Righton Biotechnology Co., Ltd, Shanghai, China
| | - Chunxia Su
- Department of Oncology, Department of Clinical Research Center, Shanghai Pulmonary Hospital &, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, 200043, China.
| |
Collapse
|
33
|
Zhao S, Gong H, Liang W. Characterization of platelet-related genes and constructing signature combined with immune-related genes for predicting outcomes and immunotherapy response in lung squamous cell carcinoma. Aging (Albany NY) 2023; 15:6969-6992. [PMID: 37477536 PMCID: PMC10415560 DOI: 10.18632/aging.204886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/26/2023] [Indexed: 07/22/2023]
Abstract
Lung squamous cell carcinoma (LUSC) is a highly malignant subtype of non-small cell lung cancer with poor prognosis. Platelets are known to play a critical role in cancer development and progression, and recent studies suggest that they can also regulate immune response in tumors. However, the relationship between platelet-related genes (PRGs) and LUSC prognosis and tumor microenvironments remains unclear. In this study, we used multiple bioinformatics algorithms to identify 25 dysregulated PRGs that were significantly associated with LUSC prognosis. We found that PRGs were involved in multiple biological processes, particularly in the tumor microenvironment, and that platelet-related scores (PRS) were a risk factor. Additionally, we established a 6-gene prognostic signature combining PRGs and immune-related genes that accurately predicted outcomes and immunotherapy efficacy in LUSC patients. Our study provides a comprehensive analysis of the biological functions and potential therapeutic targets of PRGs in LUSC, which may inform the development of new treatments for this disease.
Collapse
Affiliation(s)
- Siyi Zhao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University and Guangzhou Institute of Respiratory Disease and China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Clinical Medicine, The First Clinical Medical School of Guangzhou Medical University, Guangzhou, China
| | - Han Gong
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University and Guangzhou Institute of Respiratory Disease and China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University and Guangzhou Institute of Respiratory Disease and China State Key Laboratory of Respiratory Disease and National Clinical Research Center for Respiratory Disease, Guangzhou, China
| |
Collapse
|
34
|
Tang Y, Qian C. Research progress in leveraging biomaterials for enhancing NK cell immunotherapy. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:267-278. [PMID: 37476938 PMCID: PMC10409897 DOI: 10.3724/zdxbyxb-2022-0728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/09/2023] [Indexed: 07/22/2023]
Abstract
NK cell immunotherapy is a promising antitumor therapeutic modality after the development of T cell immunotherapy. Structural modification of NK cells with biomaterials may provide a precise, efficient, and low-cost strategy to enhance NK cell immunotherapy. The biomaterial modification of NK cells can be divided into two strategies: surface engineering with biomaterials and intracellular modification. The surface engineering strategies include hydrophobic interaction of lipids, receptor-ligand interaction between membrane proteins, covalent binding to amino acid residues, click reaction and electrostatic interaction. The intracellular modification strategies are based on manipulation by nanotechnology using membranous materials from various sources of NK cells (such as exosome, vesicle and cytomembranes). Finally, the biomaterials-based strategies regulate the recruitment, recognition and cytotoxicity of NK cells in the solid tumor site in situ to boost the activity of NK cells in the tumor. This article reviews the recent research progress in enhancing NK cell therapy based on biomaterial modification, to provide a reference for further researches on engineering NK cell therapy with biomaterials.
Collapse
Affiliation(s)
- Yingqi Tang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, State Key Laboratory of Natural Medicines, Nanjing 210009, China.
| | - Chenggen Qian
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, State Key Laboratory of Natural Medicines, Nanjing 210009, China.
| |
Collapse
|
35
|
Zhang X, Yu S, Li X, Wen X, Liu S, Zu R, Ren H, Li T, Yang C, Luo H. Research progress on the interaction between oxidative stress and platelets: Another avenue for cancer? Pharmacol Res 2023; 191:106777. [PMID: 37080257 DOI: 10.1016/j.phrs.2023.106777] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 04/22/2023]
Abstract
Oxidative stress (OS) is a chemical imbalance between an oxidant and an antioxidant, causing damage to redox signaling and control or causing molecular damage. Unbalanced oxidative metabolism can produce excessive reactive oxygen species (ROS). These excess ROS can cause drastic changes in platelet metabolism and further affect platelet function. It will also lead to an increase in platelet procoagulant phenotype and cell apoptosis, which will increase the risk of thrombosis. The creation of ROS and subsequent platelet activation, adhesion, and recruitment are then further encouraged in an auto-amplifying loop by ROS produced from platelets. Meanwhile, cancer cells produce a higher concentration of ROS due to their fast metabolism and high proliferation rate. However, excessive ROS can result in damage to and modification of cellular macromolecules. The formation of cancer and its progression is strongly associated with oxidative stress and the resulting oxidative damage. In addition, platelets are an important part of the tumor microenvironment, and there is a significant cross-communication between platelets and cancer cells. Cancer cells alter the activation status of platelets, their RNA spectrum, proteome, and other properties. The "cloaking" of cancer cells by platelets providing physical protection,avoiding destruction from shear stress and the attack of immune cells, promoting tumor cell invasion.We explored the vicious circle interaction between ROS, platelets, and cancer in this review, and we believe that ROS can play a stimulative role in tumor growth and metastasis through platelets.
Collapse
Affiliation(s)
- Xingmei Zhang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Sisi Yu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Xiaobo Li
- Molecular Diagnostic Laboratory of Department of Microbiology and Immunology, 3201 Hospital Affiliated to Medical College of Xi'an Jiaotong University, Hanzhong 723099, China
| | - Xiaoxia Wen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Shan Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Ruiling Zu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Hanxiao Ren
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Chaoguo Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China.
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China.
| |
Collapse
|
36
|
Li C, Wu J, Jiang L, Zhang L, Huang J, Tian Y, Zhao Y, Liu X, Xia L, E H, Gao P, Hou L, Yang M, Ma M, Su C, Zhang H, Chen H, She Y, Xie D, Luo Q, Chen C. The predictive value of inflammatory biomarkers for major pathological response in non-small cell lung cancer patients receiving neoadjuvant chemoimmunotherapy and its association with the immune-related tumor microenvironment: a multi-center study. Cancer Immunol Immunother 2023; 72:783-794. [PMID: 36056951 DOI: 10.1007/s00262-022-03262-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Inflammatory biomarkers in the peripheral blood have been established as predictors for immunotherapeutic efficacy in advanced non-small cell lung cancer (NSCLC). Whether they can also predict major pathological response (MPR) in neoadjuvant setting remains unclear. METHODS In this multi-center retrospective study, 122 and 92 stage I-IIIB NSCLC patients from six hospitals who received neoadjuvant chemoimmunotherapy followed by surgery were included in the discovery and external validation cohort, respectively. Baseline and on-treatment neutrophil-to-lymphocyte ratio (NLR), derived NLR (dNLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR) and systemic immune-inflammation index (SII) were calculated and associated with MPR. Furthermore, resected tumor samples from 37 patients were collected for RNA-sequencing to investigate the immune-related tumor microenvironment. RESULTS In both the discovery and validation cohorts, the on-treatment NLR, dNLR, PLR, and SII levels were significantly lower in the patients with MPR versus non-MPR. On-treatment SII remained an independent predictor of MPR in multivariate logistic regression analysis. The area under the curve (AUC) of on-treatment SII for predicting MPR was 0.75 (95%CI, 0.67-0.84) in the discovery cohort. Moreover, the predictive value was further improved by combining the on-treatment SII and radiological tumor regression data, demonstrating an AUC of 0.82 (95%CI, 0.74-0.90). The predictive accuracy was validated in the external cohort. Compared with the SII-high group, patients with SII-Low were associated with the activated B cell receptor signaling pathway and a higher intratumoral immune cell infiltration level. CONCLUSIONS On-treatment SII was independently associated with MPR in NSCLC patients receiving neoadjuvant chemoimmunotherapy. Further prospective studies are warranted.
Collapse
Affiliation(s)
- Chongwu Li
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Junqi Wu
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Long Jiang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lei Zhang
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Jia Huang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yu Tian
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yue Zhao
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Xiucheng Liu
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Lang Xia
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Haoran E
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Peigen Gao
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Likun Hou
- Department of Pathology, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Minglei Yang
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Chinese Academy of Sciences, Zhejiang, People's Republic of China
| | - Minjie Ma
- Department of Thoracic Surgery, The First Hospital of Lanzhou University, Gansu, People's Republic of China
| | - Chunxia Su
- Department of Oncology, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Hao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Xuzhou Medical University, Jiangsu, People's Republic of China
| | - Hezhong Chen
- Department of Thoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Yunlang She
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Dong Xie
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China.
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Chang Chen
- Department of Thoracic Surgery, School of Medicine, Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
37
|
Aumolertinib effectively reduces clinical symptoms of an EGFR L858R-mutant non-small cell lung cancer case coupled with osimertinib-induced severe thrombocytopenia: a case report. Anticancer Drugs 2023; 34:455-459. [PMID: 36730569 DOI: 10.1097/cad.0000000000001424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Replacement of first-generation or second-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) with third-generation EGFR-TKIs remains the current standard of care for T790M mutations in patients with non-small cell lung cancer. Osimertinib is one of the first third-generation EGFR-TKIs to be approved and is also the most widely studied in clinical research. There has been widespread concern about the adverse effects of osimertinib such as cardiotoxicity and interstitial lung disease, but few articles have reported severe thrombocytopenia after osimertinib treatment. This article reports a 64-year-old woman with non-small cell lung cancer initially diagnosed with cT2aN1M1a, EGFR p.L858R, who developed disease progression and T790M after 32 months of first-line treatment with gefitinib (250 mg/day) before switching to second-line treatment with osimertinib (80 mg/day). Severe thrombocytopenia and active bleeding occurred after treatment with osimertinib, which improved with recombinant human thrombopoietin and platelet transfusion. Treatment was replaced with aumolertinib (110 mg/day). After platelet stabilization with aumolertinib treatment in combination with chest radiotherapy, this patient had progression-free survival for 9 months and overall survival for over 45 months. In conclusion, from our experience, aumolertinib has good efficacy and mild adverse effects, and is a good choice for non-small cell lung cancer patients with T790M, especially for patients at high risk of thrombocytopenia.
Collapse
|
38
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
39
|
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: 2.0] [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
|
40
|
Magalhães-Gama F, Alves-Hanna FS, Araújo ND, Barros MS, Silva FS, Catão CLS, Moraes JS, Freitas IC, Tarragô AM, Malheiro A, Teixeira-Carvalho A, Costa AG. The Yin-Yang of myeloid cells in the leukemic microenvironment: Immunological role and clinical implications. Front Immunol 2022; 13:1071188. [PMID: 36532078 PMCID: PMC9751477 DOI: 10.3389/fimmu.2022.1071188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 11/14/2022] [Indexed: 12/02/2022] Open
Abstract
The leukemic microenvironment has a high diversity of immune cells that are phenotypically and functionally distinct. However, our understanding of the biology, immunology, and clinical implications underlying these cells remains poorly investigated. Among the resident immune cells that can infiltrate the leukemic microenvironment are myeloid cells, which correspond to a heterogeneous cell group of the innate immune system. They encompass populations of neutrophils, macrophages, and myeloid-derived suppressor cells (MDSCs). These cells can be abundant in different tissues and, in the leukemic microenvironment, are associated with the clinical outcome of the patient, acting dichotomously to contribute to leukemic progression or stimulate antitumor immune responses. In this review, we detail the current evidence and the many mechanisms that indicate that the activation of different myeloid cell populations may contribute to immunosuppression, survival, or metastatic dissemination, as well as in immunosurveillance and stimulation of specific cytotoxic responses. Furthermore, we broadly discuss the interactions of tumor-associated neutrophils and macrophages (TANs and TAMs, respectively) and MDSCs in the leukemic microenvironment. Finally, we provide new perspectives on the potential of myeloid cell subpopulations as predictive biomarkers of therapeutical response, as well as potential targets in the chemoimmunotherapy of leukemias due to their dual Yin-Yang roles in leukemia.
Collapse
Affiliation(s)
- Fábio Magalhães-Gama
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Instituto René Rachou – FIOCRUZ Minas, Belo Horizonte, Brazil
| | - Fabíola Silva Alves-Hanna
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Nilberto Dias Araújo
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Mateus Souza Barros
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Flavio Souza Silva
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
| | - Claudio Lucas Santos Catão
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Júlia Santos Moraes
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Izabela Cabral Freitas
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
| | - Andréa Monteiro Tarragô
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Adriana Malheiro
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
| | - Andréa Teixeira-Carvalho
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Instituto René Rachou – FIOCRUZ Minas, Belo Horizonte, Brazil
| | - Allyson Guimarães Costa
- Diretoria de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Instituto René Rachou - Fundação Oswaldo Cruz (FIOCRUZ) Minas, Belo Horizonte, Brazil
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Instituto de Ciências Biológicas, Universidade Federal do Amazonas (UFAM), Manaus, Brazil
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus, Brazil
- Escola de Enfermagem de Manaus, UFAM, Manaus, Brazil
| |
Collapse
|
41
|
Banna GL, Friedlaender A, Tagliamento M, Mollica V, Cortellini A, Rebuzzi SE, Prelaj A, Naqash AR, Auclin E, Garetto L, Mezquita L, Addeo A. Biological Rationale for Peripheral Blood Cell-Derived Inflammatory Indices and Related Prognostic Scores in Patients with Advanced Non-Small-Cell Lung Cancer. Curr Oncol Rep 2022; 24:1851-1862. [PMID: 36255605 DOI: 10.1007/s11912-022-01335-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW To describe the biological rationale of peripheral blood cells (PBC)-derived inflammatory indexes and assess the related prognostic scores for patients with advanced non-small cell lung cancer (aNSCLC) treated with immune-checkpoint inhibitors (ICI). RECENT FINDINGS Inflammatory indexes based on PBC may indicate a pro-inflammatory condition affecting the immune response to cancer. The lung immune prognostic index (LIPI), consisting of derived neutrophils-to-lymphocyte ratio (NLR) and lactate dehydrogenase, is a validated prognostic tool, especially for pretreated aNSCLC patients, where the combination of NLR and PD-L1 tumour expression might also be predictive of immunotherapy benefit. In untreated high-PD-L1 aNSCLC patients, the Lung-Immune-Prognostic score (LIPS), including NLR, ECOG PS and concomitant steroids, is prognostic, and its modified version might indicate patients with favourable outcomes despite an ECOG PS of 2. NLR times platelets (i.e., SII), included in the NHS-Lung score, might improve the prognostication for combined chemoimmunotherapy. PBC-derived inflammatory indexes and related scores represent accurate, reproducible and non-expensive prognostic tools with clinical and research utility.
Collapse
Affiliation(s)
| | - Alex Friedlaender
- Department of Oncology, Clinique Générale Beaulieu, Geneva, Switzerland
- Department of Oncology, University Hospital of Geneva, Geneva, Switzerland
| | - Marco Tagliamento
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Alessio Cortellini
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Sara Elena Rebuzzi
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
| | - Arsela Prelaj
- Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
- Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Abdul Rafeh Naqash
- Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
| | - Edouard Auclin
- Medical Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France
| | - Lucia Garetto
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Alfredo Addeo
- Department of Oncology, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
42
|
Tian BW, Yang YF, Yang CC, Yan LJ, Ding ZN, Liu H, Xue JS, Dong ZR, Chen ZQ, Hong JG, Wang DX, Han CL, Mao XC, Li T. Systemic immune-inflammation index predicts prognosis of cancer immunotherapy: systemic review and meta-analysis. Immunotherapy 2022; 14:1481-1496. [PMID: 36537255 DOI: 10.2217/imt-2022-0133] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective: This meta-analysis was designed to explore the association between the systemic immune-inflammation index (SII) and the therapeutic effect of immune checkpoint inhibitors. Materials & methods: The authors retrieved relevant studies published before May 25, 2022. Hazard ratio (HR) with 95% CI was used to evaluate the relationship between SII and overall survival (OS) and progression-free survival (PFS). Results: 14 articles comprising 2721 patients were included in this study. The pooled results proved that high SII levels were closely related to poor prognosis in cancer patients receiving immune checkpoint inhibitors (OS HR = 2.40; 95% CI: 2.04-2.82; PFS HR = 1.57; 95% CI: 1.33-1.86) and that an SII value of 750 was appropriate as a cut-off value (OS HR = 2.20; 95% CI: 1.83-2.63; PFS HR = 1.54; 95% CI: 1.33-1.80). Conclusion: High SII levels (>750) may be an indicator of worse OS and PFS in cancer patients treated with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Bao-Wen Tian
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Ya-Fei Yang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Chun-Cheng Yang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Lun-Jie Yan
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zi-Niu Ding
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Hui Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Jun-Shuai Xue
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhao-Ru Dong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Zhi-Qiang Chen
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Jian-Guo Hong
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Dong-Xu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Cheng-Long Han
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Xin-Cheng Mao
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, People's Republic of China
| | - Tao Li
- Department of Hepatobiliary Surgery, Second Hospital of Shandong University, Jinan, 250012, People's Republic of China
| |
Collapse
|
43
|
Deng X, Terunuma H. Harnessing NK Cells to Control Metastasis. Vaccines (Basel) 2022; 10:vaccines10122018. [PMID: 36560427 PMCID: PMC9781233 DOI: 10.3390/vaccines10122018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/21/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
In recent years, tumor immunotherapy has produced remarkable results in tumor treatment. Nevertheless, its effects are severely limited in patients with low or absent pre-existing T cell immunity. Accordingly, metastasis remains the major cause of tumor-associated death. On the other hand, natural killer (NK) cells have the unique ability to recognize and rapidly act against tumor cells and surveil tumor cell dissemination. The role of NK cells in metastasis prevention is undisputable as an increase in the number of these cells mostly leads to a favorable prognosis. Hence, it is reasonable to consider that successful metastasis involves evasion of NK-cell-mediated immunosurveillance. Therefore, harnessing NK cells to control metastasis is promising. Circulating tumor cells (CTCs) are the seeds for distant metastasis, and the number of CTCs detected in the blood of patients with tumor is associated with a worse prognosis, whereas NK cells can eliminate highly motile CTCs especially in the blood. Here, we review the role of NK cells during metastasis, particularly the specific interactions of NK cells with CTCs, which may provide essential clues on how to harness the power of NK cells against tumor metastasis. As a result, a new way to prevent or treat metastatic tumor may be developed.
Collapse
Affiliation(s)
- Xuewen Deng
- Biotherapy Institute of Japan Inc., 2-4-8 Edagawa, Koto-ku, Tokyo 135-0051, Japan
- Correspondence: ; Tel.: +81-3-5632-6080; Fax: +81-3-5632-6083
| | - Hiroshi Terunuma
- Biotherapy Institute of Japan Inc., 2-4-8 Edagawa, Koto-ku, Tokyo 135-0051, Japan
- N2 Clinic Yotsuya, 5F 2-6 Samon-cho, Shinjuku-ku, Tokyo 160-0017, Japan
| |
Collapse
|
44
|
Cheng X, Zhang H, Hamad A, Huang H, Tsung A. Surgery-mediated tumor-promoting effects on the immune microenvironment. Semin Cancer Biol 2022; 86:408-419. [PMID: 35066156 PMCID: PMC11770836 DOI: 10.1016/j.semcancer.2022.01.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
Abstract
Surgical resection continues to be the mainstay treatment for solid cancers even though chemotherapy and immunotherapy have significantly improved patient overall survival and progression-free survival. Numerous studies have shown that surgery induces the dissemination of circulating tumor cells (CTCs) and that the resultant inflammatory response promotes occult tumor growth and the metastatic process by forming a supportive tumor microenvironment (TME). Surgery-induced platelet activation is one of the initial responses to a wound and the formation of fibrin clots can provide the scaffold for recruited inflammatory cells. Activated platelets can also shield CTCs to protect them from blood shear forces and promote CTCs evasion of immune destruction. Similarly, neutrophils are recruited to the fibrin clot and enhance cancer metastatic dissemination and progression by forming neutrophil extracellular traps (NETs). Activated macrophages are also recruited to surgical sites to facilitate the metastatic spread. More importantly, the body's response to surgical insult results in the recruitment and expansion of immunosuppressive cell populations (i.e. myeloid-derived suppressor cells and regulatory T cells) and in the suppression of natural killer (NK) cells that contribute to postoperative cancer recurrence and metastasis. In this review, we seek to provide an overview of the pro-tumorigenic mechanisms resulting from surgery's impact on these cells in the TME. Further understanding of these events will allow for the development of perioperative therapeutic strategies to prevent surgery-associated metastasis.
Collapse
Affiliation(s)
- Xiang Cheng
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hongji Zhang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Ahmad Hamad
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hai Huang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Allan Tsung
- Division of Surgical Oncology, Department of Surgery, The Ohio State University James Comprehensive Cancer Center, Columbus, OH, 43210, USA.
| |
Collapse
|
45
|
Molecular Mechanism Exploration of Autologous Blood Transfusion with RBC Surface Membrane Protein pMHC/aCD28 Combined with CD8+T Cells to Promote the Proliferation of CD8+T Cells to Inhibit the Malignant Transformation of Liver Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6102672. [PMID: 36213824 PMCID: PMC9534649 DOI: 10.1155/2022/6102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022]
Abstract
Autologous blood transfusion is an important blood protection measure. Red blood cells have a certain degree of immunogenicity and their surface membrane proteins CD28 and MHC can participate in the immune response and interact with CD8+ T cells. We build a cell model with a transwell system. The binding characteristics of RBCs and CD8+ T cells were observed with a fluorescent confocal microscope. The content of the inflammatory factor TNF-α and IFN-γ produced was analyzed by ELISA. The proliferation characteristics of CD8+ T cells were analyzed by CFSE staining, and the content of CD3+CD8+ T cells was analyzed by flow cytometry. Cell migration and invasion experiments were used to analyze the malignant metastasis ability of liver cancer cells. The expression of vimentin, E-cadherin, and β-catenin was analyzed by Western blot. We establish a liver cancer model in rats and group them for autologous blood transfusion. The content of CD3+CD8+T cells in the blood of each group of rats was analyzed by flow cytometry. Western blot was used to analyze the expression of vimentin, E-cadherin, and β-catenin in the liver tissues of rats in each group. The red blood cells in the autologous reinfusion blood and CD8+ T cells have an obvious combination. The degree of combination of the two is related to the expression of CD28 and MHC. If CD28 and MHC are expressed at the same time, the combination of the two cells will be high, the proliferation of CD8+ T cells will increase, and the expression of inflammatory factors will also increase, while the expression of the three proteins that are positively correlated with the activity of cancer cells will decrease. If only one of CD28 and MHC is normally expressed, the result is contrary to the situation where both membrane proteins are normally expressed. Our project has proved that autologous infusion of red blood cell surface membrane proteins CD28 and MHC combined with CD8+ T cells can promote the proliferation of CD8+ T cells to inhibit the malignant transformation of liver cancer.
Collapse
|
46
|
Rebuzzi SE, Signori A, Stellato M, Santini D, Maruzzo M, De Giorgi U, Pedrazzoli P, Galli L, Zucali PA, Fantinel E, Carella C, Procopio G, Milella M, Boccardo F, Fratino L, Sabbatini R, Ricotta R, Panni S, Massari F, Sorarù M, Santoni M, Cortellini A, Prati V, Soto Parra H, Atzori F, Di Napoli M, Caffo O, Messina M, Morelli F, Prati G, Nolè F, Vignani F, Cavo A, Roviello G, Llaja Obispo MA, Porta C, Buti S, Fornarini G, Banna GL. The prognostic value of baseline and early variations of peripheral blood inflammatory ratios and their cellular components in patients with metastatic renal cell carcinoma treated with nivolumab: The Δ-Meet-URO analysis. Front Oncol 2022; 12:955501. [PMID: 36212433 PMCID: PMC9541611 DOI: 10.3389/fonc.2022.955501] [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: 05/28/2022] [Accepted: 08/23/2022] [Indexed: 11/14/2022] Open
Abstract
Background Treatment choice for metastatic renal cell carcinoma (mRCC) patients is still based on baseline clinical and laboratory factors. Methods By a pre-specified analysis of the Meet-URO 15 multicentric retrospective study enrolling 571 pretreated mRCC patients receiving nivolumab, baseline and early dynamic variations (Δ) of neutrophil, lymphocyte, and platelet absolute cell counts (ACC) and their inflammatory ratios (IR) were evaluated alongside their association with the best disease response and overall (OS) and progression-free survival (PFS). Multivariable analyses on OS and PFS between baseline and Δ ACC and IR values were investigated with receiving operating curves-based cut-offs. Results The analysis included 422 mRCC patients. Neutrophil-to-lymphocyte ratio (NLR) increased over time due to consistent neutrophil increase (p < 0.001). Higher baseline platelets (p = 0.044) and lower lymphocytes (p = 0.018), increasing neutrophil Δ (p for time-group interaction <0.001), higher baseline IR values (NLR: p = 0.012, SII: p = 0.003, PLR: p = 0.003), increasing NLR and systemic immune-inflammatory index (SII) (i.e., NLR x platelets) Δ (p for interaction time-group = 0.0053 and 0.0435, respectively) were associated with disease progression. OS and PFS were significantly shorter in patients with baseline lower lymphocytes (p < 0.001 for both) and higher platelets (p = 0.004 and p < 0.001, respectively) alongside early neutrophils Δ (p = 0.046 and p = 0.033, respectively). Early neutrophils and NLR Δ were independent prognostic factors for both OS (p = 0.014 and p = 0.011, respectively) and PFS (p = 0.023 and p = 0.001, respectively), alongside baseline NLR (p < 0.001 for both) and other known prognostic variables. Conclusions Early neutrophils and NLR Δ may represent new dynamic prognostic factors with clinical utility for on-treatment decisions.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy
| | - Alessio Signori
- Department of Health Sciences, Section of Biostatistics, University of Genova, Genova, Italy
| | - Marco Stellato
- SS Oncologia Medica Genitourinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Daniele Santini
- Department of Medical Oncology, Università Campus Bio-Medico of Roma, Rome, Italy
| | - Marco Maruzzo
- Oncology Unit 1, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Paolo Pedrazzoli
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy
- Medical Oncology Unit, IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Galli
- Medical Oncology Unit 2, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Paolo Andrea Zucali
- Department of Biomedical Sciences, Humanitas University, Milano, Italy
- Department of Oncology, IRCCS, Humanitas Clinical and Research Center, Milano, Italy
| | - Emanuela Fantinel
- Department of Oncology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Claudia Carella
- Division of Medical Oncology, IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | - Giuseppe Procopio
- SS Oncologia Medica Genitourinaria, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Michele Milella
- Department of Oncology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Francesco Boccardo
- Academic Unit of Medical Oncology, IRCCS Ospedale Policlinico San Martino of Genova, Genova, Italy
| | - Lucia Fratino
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano CRO-IRCCS, Aviano, Italy
| | - Roberto Sabbatini
- Medical Oncology Unit, Department of Oncology and Hemathology, University Hospital of Modena, Modena, Italy
| | | | - Stefano Panni
- Medical Oncology Unit, ASSTl– Istituti Ospitalieri Cremona Hospital, Cremona, Italy
| | - Francesco Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine, S. Orsola-Malpighi University Hospital, University of Bologna, Bologna, Italy
| | | | | | - Alessio Cortellini
- Department of Surgery and Cancer, Imperial College London, Faculty of Medicine, Hammersmith Hospital, London, United Kingdom
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Veronica Prati
- Department of Medical Oncology, Ospedale Michele e Pietro Ferrero, Verduno, Italy
| | - Hector Josè Soto Parra
- Department of Oncology, Medical Oncology, University Hospital Policlinico-San Marco, Catania, Italy
| | - Francesco Atzori
- Medical Oncology Department, University Hospital, University of Cagliari, Cagliari, Italy
| | - Marilena Di Napoli
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Orazio Caffo
- Medical Oncology Department, Santa Chiara Hospital, Trento, Italy
| | - Marco Messina
- UOC Oncologia Medica, Istituto Fondazione G. Giglio, Cefalù, Italy
| | | | - Giuseppe Prati
- Department of Oncology and Advanced Technologies AUSL - IRCCS, Reggio Emilia, Italy
| | - Franco Nolè
- Medical Oncology Division of Urogenital and Head and Neck Tumors, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Francesca Vignani
- Division of Medical Oncology, Ordine Mauriziano Hospital, Torino, Italy
| | - Alessia Cavo
- Oncology Unit, Villa Scassi Hospital, Genova, Italy
| | - Giandomenico Roviello
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Firenze, Firenze, Italy
| | | | - Camillo Porta
- Interdisciplinary Department of Medicine, University of Bari “A. Moro”, Bari, Italy
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- *Correspondence: Sebastiano Buti,
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Giuseppe Luigi Banna
- Department of Oncology, Portsmouth Hospitals University NHS Trust, Portsmouth, United Kingdom
| |
Collapse
|
47
|
Litak J, Czyżewski W, Szymoniuk M, Sakwa L, Pasierb B, Litak J, Hoffman Z, Kamieniak P, Roliński J. Biological and Clinical Aspects of Metastatic Spinal Tumors. Cancers (Basel) 2022; 14:cancers14194599. [PMID: 36230523 PMCID: PMC9559304 DOI: 10.3390/cancers14194599] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Spine metastases are a common life-threatening complication of advanced-stage malignancies and often result in poor prognosis. Symptomatic spine metastases develop in the course of about 10% of malignant neoplasms. Therefore, it is essential for contemporary medicine to understand metastatic processes in order to find appropriate, targeted therapeutic options. Our literature review aimed to describe the up-to-date knowledge about the molecular pathways and biomarkers engaged in the spine’s metastatic processes. Moreover, we described current data regarding bone-targeted treatment, the emerging targeted therapies, radiotherapy, and immunotherapy used for the treatment of spine metastases. We hope that knowledge comprehensively presented in our review will contribute to the development of novel drugs targeting specific biomarkers and pathways. The more we learn about the molecular aspects of cancer metastasis, the easier it will be to look for treatment methods that will allow us to precisely kill tumor cells. Abstract Spine metastases are a common life-threatening complication of advanced-stage malignancies and often result in poor prognosis. Symptomatic spine metastases develop in the course of about 10% of malignant neoplasms. Therefore, it is essential for contemporary medicine to understand metastatic processes in order to find appropriate, targeted therapeutic options. Thanks to continuous research, there appears more and more detailed knowledge about cancer and metastasis, but these transformations are extremely complicated, e.g., due to the complexity of reactions, the variety of places where they occur, or the participation of both tumor cells and host cells in these transitions. The right target points in tumor metastasis mechanisms are still being researched; that will help us in the proper diagnosis as well as in finding the right treatment. In this literature review, we described the current knowledge about the molecular pathways and biomarkers engaged in metastatic processes involving the spine. We also presented a current bone-targeted treatment for spine metastases and the emerging therapies targeting the discussed molecular mechanisms.
Collapse
Affiliation(s)
- Jakub Litak
- Department of Clinical Immunology, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Wojciech Czyżewski
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
- Department of Didactics and Medical Simulation, Medical University of Lublin, Chodźki 4, 20-093 Lublin, Poland
| | - Michał Szymoniuk
- Student Scientific Association at the Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Leon Sakwa
- Student Scientific Society, Kazimierz Pulaski University of Technologies and Humanities in Radom, Chrobrego 27, 26-600 Radom, Poland
| | - Barbara Pasierb
- Department of Dermatology, Radom Specialist Hospital, Lekarska 4, 26-600 Radom, Poland
- Correspondence:
| | - Joanna Litak
- St. John’s Cancer Center in Lublin, Jaczewskiego 7, 20-090 Lublin, Poland
| | - Zofia Hoffman
- Student Scientific Society, Medical University of Lublin, Al. Racławickie 1, 20-059 Lublin, Poland
| | - Piotr Kamieniak
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Jacek Roliński
- Department of Clinical Immunology, Medical University of Lublin, Chodźki 4A, 20-093 Lublin, Poland
| |
Collapse
|
48
|
Investigation of Biomarkers Associated with Low Platelet Counts in Normal Karyotype Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms23147772. [PMID: 35887121 PMCID: PMC9320053 DOI: 10.3390/ijms23147772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) patients are at risk of bleeding due to disease-related lack of platelets and systemic coagulopathy. Platelets play a role in hemostasis. Leukemic blasts have been shown to alter platelet activation in vitro. Here we investigated biomarkers associated with thrombocytopenia in normal karyotype AML (NK-AML). From The Cancer Genome Atlas database, case-control study was performed between normal karyotype (NK) platelet-decreased AML (PD-AML, platelet count < 100 × 109/L, n = 24) and NK platelet-not-decreased AML (PND-AML, with platelet count ≥ 100 × 109/L, n = 13). Differentially expressed gene analysis, pathway analysis and modelling for predicting platelet decrease in AML were performed. DEG analysis and pathway analysis revealed 157 genes and eight pathways specific for PD-AML, respectively. Most of the eight pathways were significantly involved in G-protein-coupled receptor-related pathway, cytokine-related pathway, and bone remodeling pathway. Among the key genes involved in at least one pathway, three genes including CSF1R, TNFSF15 and CLEC10A were selected as promising biomarkers for predicting PD-AML (0.847 of AUC in support vector machine model). This is the first study that identified biomarkers using RNA expression data analysis and could help understand the pathophysiology in AML with low platelet count.
Collapse
|
49
|
Wang P, Zhao W, Cao H. Development of a Platelet-Related Prognostic Model for Colorectal Cancer. Front Genet 2022; 13:904168. [PMID: 35719389 PMCID: PMC9198283 DOI: 10.3389/fgene.2022.904168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 04/26/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) represents one of the most common malignancies with high morbidity worldwide. Growing evidence has suggested that platelets are a fundamental component of the tumor microenvironment and play crucial roles in driving tumor biological behavior. The construction of a platelet-related prognostic model that can reliably predict CRC prognosis is of great clinical significance. The 1427 CRC-specific platelet-related genes were collected and mainly enriched in the ribosome and immune-related pathways. Based on platelet-related genes, three subtypes of TCGA CRC samples were identified by consensus clustering and characterized by differences in angiogenesis, epithelial–mesenchymal transition, immune infiltration, and prognosis. A total of 100 prognostic platelet-related genes were identified by univariate Cox regression. LASSO Cox regression further shrank those genes and constructed a 10-gene prognostic model. The patients with higher risk scores had significantly worse disease-specific survival than those with lower scores in both TCGA and validation cohorts. The risk score demonstrated good predictive performance for prognosis by receiver operating characteristic (ROC) curves. Furthermore, multivariate Cox regression analysis showed that the risk score was independent of TNM stage, sex, and age, and a graphic nomogram based on the risk score and clinical factors was developed to predict survival probability of CRC patients. Patients from the high-risk group were characterized by higher infiltration of immunosuppressive cells such as MDSC and Treg and higher expression of checkpoints CTLA4, CD86, and PDCD1LG2. Taken together, we identified three platelet-related subtypes and specifically constructed a promising 10-gene prognostic model in CRC. Our results highlighted the potential survival effects of platelet-related genes and provided evidence about their roles in regulating tumor immunity.
Collapse
Affiliation(s)
- Pengcheng Wang
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital, Taiyuan, China
| | - Wei Zhao
- Department of Anesthesiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hailei Cao
- Department of Colorectal and Anal Surgery, Shanxi Province Cancer Hospital, Taiyuan, China
| |
Collapse
|
50
|
Marini I, Uzun G, Jamal K, Bakchoul T. Treatment of drug-induced immune thrombocytopenias. Haematologica 2022; 107:1264-1277. [PMID: 35642486 PMCID: PMC9152960 DOI: 10.3324/haematol.2021.279484] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Indexed: 01/19/2023] Open
Abstract
Several therapeutic agents can cause thrombocytopenia by either immune-mediated or non-immune-mediated mechanisms. Non-immune-mediated thrombocytopenia is due to direct toxicity of drug molecules to platelets or megakaryocytes. Immune-mediated thrombocytopenia, on the other hand, involves the formation of antibodies that react to platelet-specific glycoprotein complexes, as in classic drug-induced immune thrombocytopenia (DITP), or to platelet factor 4, as in heparin-induced thrombocytopenia (HIT) and vaccine-induced immune thrombotic thrombocytopenia (VITT). Clinical signs include a rapid drop in platelet count, bleeding or thrombosis. Since the patient's condition can deteriorate rapidly, prompt diagnosis and management are critical. However, the necessary diagnostic tests are only available in specialized laboratories. Therefore, the most demanding step in treatment is to identify the agent responsible for thrombocytopenia, which often proves difficult because many patients are taking multiple medications and have comorbidities that can themselves also cause thrombocytopenia. While DITP is commonly associated with an increased risk of bleeding, HIT and VITT have a high mortality rate due to the high incidence of thromboembolic complications. A structured approach to drug-associated thrombocytopenia/thrombosis can lead to successful treatment and a lower mortality rate. In addition to describing the treatment of DITP, HIT, VITT, and vaccine-associated immune thrombocytopenia, this review also provides the pathophysiological and clinical information necessary for correct patient management.
Collapse
Affiliation(s)
- Irene Marini
- Centre for Clinical Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen
| | - Gunalp Uzun
- Centre for Clinical Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen
| | - Kinan Jamal
- Centre for Clinical Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen
| | - Tamam Bakchoul
- Centre for Clinical Transfusion Medicine, Medical Faculty of Tübingen, University of Tübingen.
| |
Collapse
|