1
|
Baskaran D, Liu Y, Zhou J, Wang Y, Nguyen D, Wang H. In vitro and in vivo metabolic tagging and modulation of platelets. Mater Today Bio 2025; 32:101719. [PMID: 40236816 PMCID: PMC11999579 DOI: 10.1016/j.mtbio.2025.101719] [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: 12/31/2024] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Platelets play a critical role in hemostasis at sites of injury and are capable of interacting with various types of cells in the bloodstream. The promise of utilizing platelets for diagnostic and therapeutic applications has motivated the development of facile strategies to functionalize platelets. However, platelets with a small size, lack of nucleus and efficient protein machinery, and low tolerance to chemicals and transfection agents have posed significant challenges for chemical or genetic engineering. Here, for the first time, we report successful metabolic glycan labeling of platelets to introduce chemical tags (e.g., azido groups) onto the membrane of platelets. We demonstrate that azido-sugars can metabolically label platelets in a concentration dependent manner, with cell-surface azido groups detectable at as early as 4 hours. The cell-surface azido groups enable the conjugation of various macromolecular cargos including proteins and polymers onto platelets via efficient click chemistry. Small-molecule drugs such as doxorubicin can also be conjugated onto azido-labeled platelets and become subsequently released to kill surrounding cancer cells, demonstrating the feasibility of utilizing platelets as a drug delivery vehicle. We further show that azido-sugars, upon intraperitoneal injection, can metabolically label platelets with azido groups in vivo, which persist for up to 4 days in mice (nearly the life-span of murine platelets). This in vitro and in vivo platelet labeling and targeting technology opens a new avenue to platelet-based diagnostics and therapeutics.
Collapse
Affiliation(s)
- Dhyanesh Baskaran
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Daniel Nguyen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
2
|
Hussain MS, Moglad E, Goyal A, Rekha MM, Sharma GC, Jayabalan K, Sahoo S, Devi A, Goyal K, Gupta G, Shahwan M, Alzarea SI, Kazmi I. Tumor-educated platelets in lung cancer. Clin Chim Acta 2025; 573:120307. [PMID: 40228574 DOI: 10.1016/j.cca.2025.120307] [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: 03/24/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Non-invasive diagnostic monitoring techniques have become essential for treating lung cancer (LC), which continues to be the primary cause of cancer-related death worldwide. The new diagnostic biomarkers called tumour-educated platelets (TEPs) show strong prospects for providing vital information about tumor biology, tumor spread pathways, and treatment reaction patterns. Despite lacking a nucleus, platelets exhibit an active RNA profile that develops through interactions with tumor-derived compounds and the tumor microenvironments (TME). This review explains platelet-tumour interaction regulatory mechanisms while focusing on platelet contributions toward cancer development, immune system avoidance, and blood clot formation. The detection and classification of LC show promise through the analysis of RNA molecules extracted from platelets that encompass mRNAs and non-coding RNAs. RNA sequencing technology based on TEP demonstrates excellent diagnostic power by correctly identifying LC patients alongside their oncogenic alterations of EGFR, KRAS, and ALK. Treatment predictions have proven successful using platelet RNA profiles, specifically in immunotherapy and targeted therapy. Integrating next-generation sequencing with machine learning and artificial intelligence enhances TEP-based diagnostic tools, improving detection accuracy. Standardizing platelet extraction methods and vesicle purification from tumor material needs better development for effective and affordable clinical use. Future investigations should combine TEPs with circulating tumor DNA and exosomal RNA markers to enhance both earliest-stage LC diagnosis and patient-specific therapeutic approaches. TEPs introduce a groundbreaking technique in oncology since they can transform non-invasive medical diagnostics and therapeutic monitoring for cancer.
Collapse
Affiliation(s)
- Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Alkharj 11942, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Karthikeyan Jayabalan
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Samir Sahoo
- Department of General Medicine IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf 72341, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
3
|
Zhang S, Zhu Z, Liu L, Nashan B, Zhang S. Biomarker, efficacy and safety analysis of transcatheter arterial chemoembolization combined with atezolizumab and bevacizumab for unresectable hepatocellular carcinoma. Cancer Immunol Immunother 2025; 74:209. [PMID: 40387956 PMCID: PMC12089556 DOI: 10.1007/s00262-025-04058-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE Transcatheter arterial chemoembolization (TACE) combined with atezolizumab and bevacizumab (Atezo-Bev) [Atezo-Bev-TACE] has shown promising therapeutic efficacy in patients with unresectable hepatocellular carcinoma (uHCC). However, there is currently no published research on biomarkers that can predict the treatment outcomes of Atezo-Bev-TACE. This study aims to evaluate the predictive value of the baseline neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in uHCC patients undergoing Atezo-Bev-TACE treatment. METHODS This retrospective study included uHCC patients who received Atezo-Bev-TACE and tyrosine kinase inhibitors (TKIs) at the First Affiliated Hospital of the University of Science and Technology of China between November 1, 2020, and November 1, 2023. The primary endpoint of the study was the correlation between baseline NLR and PLR with overall survival (OS) and progression-free survival (PFS). The secondary endpoints were the efficacy and safety of the Atezo-Bev-TACE regimen. RESULTS Among the 71 enrolled patients with uHCC who received Atezo-Bev-TACE therapy, the objective response rate was 55.0%, with a median OS of 20.0 months (95% confidence interval [CI] 17.4-21.0 months) and a median PFS of 10.4 months (95% CI 7.7-13.1 months). Patients with tumor response had significantly lower baseline NLR and PLR values compared to those without response (2.5 vs. 4.0, P < 0.001; 106.9 vs. 131.3, P = 0.001). The optimal cut-off values for NLR and PLR were determined to be 2.9 and 148.0, respectively, based on receiver operating characteristic curves. Patients with baseline NLR < 2.9 had significantly longer median OS (not reached vs. 17.8 months, P = 0.014) and improved median PFS (15.6 months vs. 9.3 months, P = 0.034) compared to those with NLR ≥ 2.9. Similarly, patients with baseline PLR < 148.0 had a significantly better median OS (20.0 months vs. 12.0 months, P = 0.004) and longer median PFS (13.7 months vs. 6.4 months, P < 0.001) compared to those with PLR ≥ 148.0. Univariate and multivariate Cox regression analyses identified baseline PLR ≥ 148.0 as an independent risk factor for poorer survival outcomes. Additionally, most adverse events (AEs) observed during Atezo-Bev-TACE treatment were grade 1-2, with fewer grade 3-4 AEs, and no grade 5 AEs were reported. Comparative analysis between the Atezo-Bev-TACE group (71 patients) and the TKIs-TACE group (63 patients) demonstrated that the ORR of the TKIs-TACE group was 34.9%, lower than that of the Atezo-Bev-TACE group (55.0%). No statistically significant differences were observed in baseline characteristics between the two groups before treatment. The median OS in the Atezo-Bev-TACE group was 20.0 months, significantly superior to the 14.7 months in the TKIs-TACE group (P = 0.005). Similarly, the median PFS in the Atezo-Bev-TACE group was 10.4 months, significantly better than the 7.8 months in the TKIs-TACE group (P = 0.008). CONCLUSION A baseline NLR ≥ 2.9 and PLR ≥ 148.0 may serve as predictive factors for poor OS and PFS in uHCC patients receiving Atezo-Bev-TACE treatment. Furthermore, the Atezo-Bev-TACE regimen demonstrates good efficacy and safety in the clinical management of uHCC patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/drug therapy
- Liver Neoplasms/therapy
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/blood
- Male
- Female
- Middle Aged
- Retrospective Studies
- Bevacizumab/therapeutic use
- Bevacizumab/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Chemoembolization, Therapeutic/methods
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor
- Neutrophils
- Adult
- Treatment Outcome
Collapse
Affiliation(s)
- Shaobo Zhang
- Department of Liver Transplantation, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Zebin Zhu
- Department of Liver Transplantation, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Lianxin Liu
- Department of Liver Transplantation, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China
| | - Björn Nashan
- Department of Liver Transplantation, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China.
| | - Shugeng Zhang
- Department of Liver Transplantation, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230000, Anhui, China.
| |
Collapse
|
4
|
Yuan Z, Guo L, Pang Y, Wang W, Shang Y, Xie C, Qian C, Sun J, Wu X. Activated platelets stimulate effector CD8+ T cells to enhance HNSCC immunotherapy efficacy. Discov Oncol 2025; 16:760. [PMID: 40366505 PMCID: PMC12078921 DOI: 10.1007/s12672-025-02596-y] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
Programmed Death Receptor-1 (PD-1) is an immune checkpoint receptor expressed on the surface of T cells. Monoclonal antibodies targeting PD-1 and its ligand, PD-L1, are among the most widely utilized immune checkpoint inhibitors in cancer immunotherapy, dramatically improving the prognosis of patients with various malignancies. Traditionally, platelets, which are cytoplasmic fragments derived from megakaryocytes, have been primarily recognized for their roles in hemostasis and coagulation. However, recent studies have highlighted the emerging role of platelets in cancer biology and therapy. Platelets can modulate immune cell functions through various mechanisms, including the release of bioactive molecules and direct interactions with immune cells. A deeper understanding of the interplay between platelets and immune responses could pave the way for novel therapeutic strategies in cancer treatment. In our research, in patients with better treatment responses, there are higher levels of mature and activated CD8+ T cells in their PBMCs prior to treatment. Additionally, the activation of platelets is also more pronounced, and the proteins expressed on these platelets may modulate immune cells. After receiving immunotherapy, patients in the responsive (R) group exhibited a higher abundance of activated effector CD8+ T cells, which demonstrated stronger immune response capabilities. Furthermore, the increased levels of activated platelets in the R group may contribute to the regulation of CD8+ effector memory T cells, influencing their quantity and function. Our study suggests that the functional state of CD8+ T cells and the level of activated platelets prior to treatment may serve as predictive indicators for the efficacy of PD-1 inhibitors in head and neck cancer patients. Activated CD8+ effector T cells may contribute to the differences in immunotherapy responses, with activated platelets playing a role in promoting the maturation and activation of CD8+ effector memory T cells.These insights help better understand the interactions between platelets and immune cells, particularly emphasizing the role of CD8+ effector memory T cells in immunotherapy. Additionally, they offer potential strategies for predicting patient responses to PD-1 inhibitor treatment and optimizing the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Lunhua Guo
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Yuheng Pang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Yuefeng Shang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Chufei Xie
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Cheng Qian
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China.
| | - Ji Sun
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Haping RD NO.150, Harbin, 150001, Heilongjiang, People's Republic of China.
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, The Fourth Affiliated Hospital of Harbin Medical University, No. 37 Yiyuan Street, Nangang District, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
5
|
Lu Q, Han Z, Wang X, Du L, Fan X, Zhao J, Zhu R, Wang H, Song J, Shen W, Zhang H, He Z, Wang K, Sun J. Long-acting bioengineered platelets with internal doxorubicin loaded and external quercetin liposomes anchored for post-surgical tumor therapy. J Control Release 2025; 381:113546. [PMID: 39983926 DOI: 10.1016/j.jconrel.2025.02.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/10/2025] [Accepted: 02/17/2025] [Indexed: 02/23/2025]
Abstract
Due to its natural tumor targeting ability, platelet-based drug delivery platform shows the great potential for tumor targeted treatment. However, both limited in vitro storage stability and rapid in vivo clearance rate severely restrict its clinical application. Here, utilizing the spatial structure of platelets precisely, chemotherapy drug doxorubicin (Dox) and liposomes-containing quercetin (Que) are loaded inside and anchored outside platelets, respectively, for establishing the engineered platelet platform (PDQLs). Dox plays the important role in inhibiting tumor growth, while Que mainly inhibits platelet apoptosis through activating serine/threonine protein kinase. PDQLs show the strong ability to resist external stimulation and physical damage. After being stored at room temperature for 4 days, more than 70 % of the platelets remain active. Given the natural wound tropism and tumor targeting abilities, the tumor accumulation of PDQLs is 1.02-fold higher than that of the solution. Base on the stealth characteristics of platelets and the continuous action of Que, PDQLs exhibit 10.63-fold increase area under the curve of solution. PDQLs can balance the anti-tumor recurrence and metastasis efficacy after surgery and safety. Our findings open a promising perspective and new sights for the development of bioengineered platelet platform in clinical application.
Collapse
Affiliation(s)
- Qi Lu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zeyu Han
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xia Wang
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Lili Du
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Xiaoyuan Fan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Jian Zhao
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Ruihong Zhu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Helin Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Jiaxuan Song
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Wenwen Shen
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Haotian Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China
| | - Kaiyuan Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China.
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, PR China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, PR China.
| |
Collapse
|
6
|
Yang Y, Liu X, Zhang R, Liu Y, Zhou N, Jiang Y. Size-Tunable Micro-Nano Liposomes: Enhanced Lung Targeting and Tumor Penetration for Combination Treatment of Lung Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409593. [PMID: 40237096 DOI: 10.1002/smll.202409593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 03/26/2025] [Indexed: 04/17/2025]
Abstract
The inefficient delivery of nanocarriers and drug resistance seriously limit therapeutic effects of lung cancer. Here, a size-tunable micro-nano liposome system, PCAL@TM, is designed for targeted delivery of paclitaxel (PTX) and oxygen to lung tumors. PTX-loaded corosolic acid (CA) nano-liposomes (PCAL, 100 nm) are anchored to the surface of oxygenated perfluorotributylamine (TBA)-loaded multivesicular liposomes (TM, 10 µm) via the biotin-avidin interactions with matrix metalloproteinase-9 (MMP-9) cleavable linker. After intravenous administration to lung tumor-bearing mice, the distribution amount of PCAL@TM in the lungs is extremely higher than that in the liver and spleen. The MMP-9-sensitive PCAL@TM can decouple into nano-PCAL and micro-TM in tumors; while, TMs enable breaking into smaller vesicles under vascular pressure, and release oxygen leading to the downregulation of HIF-1α and platelet-activated TGF-β. Meanwhile, PCAL can penetrate deeply into tumor by the tumor-targeted-penetrable CA liposomes, to promote the reduction of inflammation levels and enhance PTX-induced immunogenic cell death (ICD). Together, these results lead to the reversals of chemoresistance and tumor immunosuppressive, achieving significant improvement in PTX chemotherapy and α-PD-1 immunotherapy. The PCAL@TM system presents a novel strategy to enhance the efficiency of nano-drug delivery and the outcome of combined therapy for lung tumor.
Collapse
Affiliation(s)
- Yueying Yang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xiao Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Ruizhe Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yunhu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Nan Zhou
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yanyan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education (Fudan University), Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
7
|
Wang T, Chen L, Bao X, Han Z, Wang Z, Nie S, Gu Y, Gong J. Short-term peri- and intra-tumoral CT radiomics to predict immunotherapy response in advanced non-small cell lung cancer. Transl Lung Cancer Res 2025; 14:785-797. [PMID: 40248738 PMCID: PMC12000948 DOI: 10.21037/tlcr-24-973] [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: 10/21/2024] [Accepted: 02/08/2025] [Indexed: 04/19/2025]
Abstract
Background Predicting response to immunotherapy is crucial for advanced non-small cell lung cancer (NSCLC) treatment planning, but effective predictive markers for immunotherapy efficacy are still lacking. This study aimed to develop an explainable machine learning model for predicting immunotherapy responses in advanced NSCLC patients. Methods A total of 245 advanced NSCLC patients from two centers who received immunotherapy were retrospectively enrolled. For each primary tumor, three regions of interest were analyzed, namely, the intratumoral region (ITR), peritumoral region (PTR), and combined intratumoral and PTR (IPTR). Pre-radiomics features and delta-radiomics features reflecting the rate of change between radiomics features before and after treatment were extracted. Models for predicting immunotherapy responses were established via the extreme gradient boosting (XGBoost) classifier and assessed in terms of discrimination, calibration, and clinical utility. The SHapley Additive exPlanations (SHAP) tool was employed to explore the interpretability of the model. Kaplan-Meier (KM) analysis of progression-free survival (PFS) was conducted to evaluate the prognostic value of the prediction models. Results The delta-radiomics models of ITR and IPTR demonstrated optimal performance in predicting immunotherapy response, significantly improving the area under the curve (AUC) to 0.85 and 0.83 in the internal validation cohort and 0.84 and 0.86 in the external validation cohort. SHAP revealed a strong relationship between the delta-radiomics feature values and the model-predicted probabilities. KM curves indicated that the high-risk groups identified by the delta-radiomics models had significantly worse PFS than did the low-risk groups across all cohorts. Conclusions The results demonstrated that a model based on multiple time points outperformed one based on a single time point. The delta-radiomics model has been proved a noninvasive approach for assessing the response of advanced NSCLC patients to immunotherapy and facilitates individualized treatment decision making.
Collapse
Affiliation(s)
- Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Chen
- Department of Radiology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiao Bao
- Department of Radiology, Shanghai Pulmonary Hospital, Shanghai, China
| | - Zijuan Han
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shengdong Nie
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Gong
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Liu J, Wang H, Wang X, Jassem J, Si J, Liu Y, Jin J. Biological and clinical significance of circulating mucosal-associated invariant T cells in lung cancer. Transl Cancer Res 2025; 14:1995-2009. [PMID: 40224974 PMCID: PMC11985213 DOI: 10.21037/tcr-2025-178] [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: 01/18/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Background Lung cancer is among the most common malignant tumors worldwide. Circulating mucosal-associated invariant T (cMAIT) cells play an important role in cancer. This study investigated the biological and clinical significance of cMAIT cells in lung cancer. Methods Fasting peripheral blood mononuclear cells (PBMCs) were extracted from 30 newly diagnosed lung cancer patients and 30 healthy controls. The percentages of cMAIT among the CD3+T cells, their absolute values, and subpopulation distribution in both groups were compared by flow cytometry. The correlations of cMAIT with the neutrophil-to-lymphocyte ratio (NLR) and the expression of programmed cell death-ligand 1 (PD-L1) were analyzed. Enzyme-linked immunosorbent assay (ELISA) was used to detect plasma interleukin-6 (IL-6), interleukin-8 (IL-8), and interferon-γ (IFN-γ) levels in lung cancer patients and healthy controls. The percentage of MAIT cells in the tumor tissues and adjacent normal lung tissues was measured by flow cytometry. Results The percentages and absolute values of the cMAIT in lung cancer patients were lower than in healthy subjects (P<0.001, P<0.01, respectively). The CD8+CD4- subgroup was dominant in both groups. There was no significant difference in percentages of the CD8+CD4- subgroup between lung cancer patients and healthy subjects (P=0.63), but the absolute values of CD8+CD4- cells were lower in lung cancer patients (P<0.05). The percentages and absolute values of cMAIT in lung cancer patients were negatively correlated with NLR (r=-0.537; P<0.01 and r=-0.423; P<0.05, respectively). The cMAIT cell percentage did not correlate with PD-L1 tumor expression (r=-0.1740; P=0.59) and with the PD-L1 expression level (P>0.99). No differences were found in the plasma IL-6, IL-8, and IFN-γ levels in lung cancer patients and healthy controls (P=0.63, P=0.052, P=0.13, respectively). The percentage of mucosal-associated invariant T (MAIT) cells in lung cancer tissues was higher than in the adjacent normal lung tissues (1.44% vs. 1.29%, P=0.044). Conclusions Lower percentage and absolute values of cMAIT in lung cancer patients may be due to their migration into tissues. The number of cMAIT in lung cancer patients may potentially be considered as a prognostic indicator.
Collapse
Affiliation(s)
- Jingjing Liu
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haoyu Wang
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaotao Wang
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Jiming Si
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanhua Liu
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianjun Jin
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Langiu M, Crescence L, Mège D, Dubois C, Panicot-Dubois L. Consequences of platelet-educated cancer cells on the expression of inflammatory and metastatic glycoproteins. PLoS One 2025; 20:e0317096. [PMID: 40096084 PMCID: PMC11913274 DOI: 10.1371/journal.pone.0317096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/20/2024] [Indexed: 03/19/2025] Open
Abstract
Cancer-associated thrombosis, a major cause of mortality in cancer patients, exhibits a 4 to 7 times higher incidence compared to the general population. Platelet activation by tumor cells contributes to this pro-thrombotic state. Cancer cell-educated platelets have also been described to be implicated in promoting metastasis. Intriguingly, our team, among others, unveils a reverse process, wherein platelets educate cancer cells by transferring lipids, RNAs, and proteins. Here, focusing on colorectal and pancreatic cancers, our study investigates genes and proteins mediating platelet education of cancer cells. We demonstrated, for the first time, that platelets can educate cancer cells by inducing changes in the transcription of genes related to glycosylation, inflammation, and metastasis in cancer cells themselves. These results indicate a direct impact of platelets on cancer cell phenotype. This novel insight suggests potential therapeutic avenues for cancer treatment, disrupting platelet-mediated alterations and influencing the course of cancer progression.
Collapse
Affiliation(s)
- Mélanie Langiu
- Aix Marseille University, INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), INSERM 1263 (Institut National de la Santé et de la Recherche), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
| | - Lydie Crescence
- Aix Marseille University, INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), INSERM 1263 (Institut National de la Santé et de la Recherche), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
| | - Diane Mège
- Aix Marseille University, INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), INSERM 1263 (Institut National de la Santé et de la Recherche), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
- Department of Digestive Surgery, La Timone University Hospital, Marseille, France
| | - Christophe Dubois
- Aix Marseille University, INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), INSERM 1263 (Institut National de la Santé et de la Recherche), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
| | - Laurence Panicot-Dubois
- Aix Marseille University, INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), INSERM 1263 (Institut National de la Santé et de la Recherche), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), Marseille, France
| |
Collapse
|
10
|
Zheng K, Zhang J, Xu T, Li F, Li F, Zeng J, Guo Y, Hao Z. Establishment and validation of a survival prediction model for stage IV non-small cell lung cancer: a real-world study. Front Immunol 2025; 16:1508721. [PMID: 40114917 PMCID: PMC11922824 DOI: 10.3389/fimmu.2025.1508721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Objective The aim of this study is to develop and validate a predictive model for predicting survival in individual advanced non-small cell lung cancer patients by integrating basic patient information and clinical data. Methods A total of 462 patients with advanced non-small cell lung cancer collected from Shanxi Cancer Hospital were randomly assigned (in a 7:3 ratio) to a training cohort and an internal validation cohort. Independent factors affecting patients' 3-year survival were screened and predictive models were created by using a single-factor followed by multifactor Cox regression analysis. Evaluate the performance of the model using the consistency index (C-index), calibration curves, receiver operating characteristic curves (ROC) and decision curve analysis (DCA). The collected patients who received chemotherapy alone and those who received chemotherapy combined with immunotherapy were statistically paired using propensity score matching between the two groups, and subgroup analyses were performed among the screened variables. Results A better prognostic model was created and a nomogram chart visualizing the model was drawn. Based on the median risk score of the training cohort, all individuals were categorized into high- and low-risk groups, with the high-risk group having worse OS in both cohorts (P<0.05). The results of subgroup analysis showed that chemotherapy alone versus chemotherapy combined with immunotherapy in patients with advanced NSCLC affected OS. Conclusion A clinical predictive model was developed to predict 3-year survival in patients with advanced non-small cell lung cancer. The study demonstrated that chemotherapy combined with immunotherapy is superior to chemotherapy alone.
Collapse
Affiliation(s)
- Keao Zheng
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Junyan Zhang
- Department of Affiliated Cancer Hospital, Shanxi Medical University, Taiyuan, China
| | - Tingting Xu
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Fangyu Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, China
| | - Feng Li
- Department of Pharmacy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jing Zeng
- Department of Pharmacy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Yimeng Guo
- Department of Pharmacy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Zhiying Hao
- Department of Pharmacy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| |
Collapse
|
11
|
Josefsson EC. Platelets and megakaryocytes in cancer. J Thromb Haemost 2025; 23:804-816. [PMID: 39742972 DOI: 10.1016/j.jtha.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Platelets have important roles in hemostasis but also actively participate in cancer metastasis and inflammatory processes. They are produced by large precursor cells, the megakaryocytes, residing mainly in the bone marrow. Clinically, elevated platelet counts and/or increased platelet-to-lymphocyte ratio are being explored as biomarkers of metastatic disease and to predict survival or response to therapy in certain cancers. Multiple mechanisms have been put forward on how platelets promote hematogenous metastasis stemming mainly from murine experimental models. Research is now beginning to explore the potential roles of megakaryocytes in solid cancer, myeloma, and lymphoma. Here, we review mechanisms on how platelets and megakaryocytes contribute to cancer progression and metastasis but also discuss potential cancer-suppressing functions mainly related to the regulation of vascular intratumor integrity. Recent developments in cancer immune checkpoint therapy are reviewed with a focus on the potential roles of platelets. Moreover, we review studies exploring platelets for targeted drug delivery systems in cancer therapy.
Collapse
Affiliation(s)
- Emma C Josefsson
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Chemistry, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, The University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
12
|
Lee SW, Jeong S, Kim YJ, Noh JE, Rho KN, Kim HO, Cho HJ, Yang DH, Hwang EC, Kyun Bae W, Yun SJ, Yun JS, Park CK, Oh IJ, Cho JH. Enhanced thrombopoiesis supplies PD-L1 to circulating immune cells via the generation of PD-L1-expressing platelets in patients with lung cancer. J Immunother Cancer 2025; 13:e010193. [PMID: 40010769 PMCID: PMC11865743 DOI: 10.1136/jitc-2024-010193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/30/2024] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND The increased expression of programmed cell death ligand 1 (PD-L1) on a subset of immune cells in the peripheral blood has been frequently observed in patients with cancer, suggesting a relationship with PD-L1 expression in tumor tissues. In this study, we investigated the mechanisms underlying PD-L1 expression on various types of immune cells in the peripheral blood of patients with cancer. METHODS PD-L1 expression on various immune cell populations was analyzed in peripheral blood mononuclear cells of 112 patients with non-small cell lung cancer (NSCLC) using flow cytometry. A mouse model of X-ray-induced acute thrombocytopenia was used to investigate the relationship between thrombopoiesis and PD-L1-expressing platelet generation. The clinical significance of PD-L1-expressing platelets was analyzed in a cohort of patients with stage IV NSCLC who received a combination of anti-programmed cell death 1 (PD-1) therapy and chemotherapy. RESULTS All immune cell populations, including monocytes, T cells, B cells, and NK cells, showed higher PD-L1 expression in patients with cancer than in healthy controls. However, this increased frequency of PD-L1-expressing cells was not attributed to the expression of the cells themselves. Instead, it was entirely dependent on the direct interaction of the cells with PD-L1-expressing platelets. Notably, the platelet-dependent acquisition of PD-L1 on circulating immune cells of patients with lung cancer was observed in various other cancer types and was mechanistically associated with a surge in thrombopoiesis, resulting in the increased production of PD-L1-expressing reticulated platelets. Clinically, patients with enhanced thrombopoiesis and concurrently high PD-L1-expressing platelets exhibited a better response to anti-PD-1 therapy. CONCLUSIONS These findings highlight the role of tumor-associated thrombopoiesis in generating PD-L1-expressing platelets that may serve as a resource for PD-L1-positive cells in the circulation and act as a predictive biomarker for anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Sung-Woo Lee
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Saei Jeong
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Young Ju Kim
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Jeong Eun Noh
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Kyung Na Rho
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Hee-Ok Kim
- Selecxine Inc, Seoul, Korea (the Republic of)
| | - Hyun-Ju Cho
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Deok Hwan Yang
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Eu Chang Hwang
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Department of Urology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Woo Kyun Bae
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Sook Jung Yun
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Department of Dermatology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Ju Sik Yun
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Thoracic and Cardiovascular Surgery, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Cheol-Kyu Park
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Chonnam National University Hwasun Hospital, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| | - Jae-Ho Cho
- Department of Microbiology and Immunology, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Hwasun-eup, Hwasun-gun, Jeollanam-do, Korea (the Republic of)
| |
Collapse
|
13
|
Zou M, Qattan A, Al-Alwan M, Ghebeh H, Binjumah N, Al-Haj L, Khabar KSA, Altaweel A, Almohanna F, Assiri AM, Aboussekhra A, Alzahrani AS, Shi Y. Genome-wide transcriptome analysis and drug target discovery reveal key genes and pathways in thyroid cancer metastasis. Front Endocrinol (Lausanne) 2025; 16:1514264. [PMID: 39996058 PMCID: PMC11847698 DOI: 10.3389/fendo.2025.1514264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Introduction Metastasis is the major cause of thyroid cancer morbidity and mortality. However, the mechanisms are still poorly understood. Methods We performed genome-wide transcriptome analysis comparing gene expression profile of metastatic thyroid cancer cells (Met) with primary tumor cells established from transgenic mouse models of papillary thyroid cancer (PTC), follicular thyroid cancer (FTC), poorly differentiated thyroid cancer (PDTC), and anaplastic thyroid cancer (ATC). Results Genes involved in tumor microenvironment (TME), inflammation, and immune escape were significantly overexpressed in Met cells. Notably, IL-6-mediated inflammatory and PD-L1 pathways were highly active in Met cells with increased secretion of pro-inflammatory and pro-metastatic cytokines such as CCL2, CCL11, IL5, IL6, and CXCL5. Furthermore, Met cells showed robust overexpression of Tbxas1, a thromboxane A synthase 1 gene that catalyzes the conversion of prostaglandin H2 to thromboxane A2 (TXA2), a potent inducer of platelet aggregation. Application of aspirin, a TXA2 inhibitor, significantly reduced lung metastases. Mertk, a member of the TAM (Tyro, Axl, Mertk) family of RTKs, was also overexpressed in Met cells, which led to increased MAPK activation, epithelial-mesenchymal transition (EMT), and enrichment of cancer stem cells. Braf-mutant Met cells developed resistance to BRAFV600E inhibitor PLX4720, but remained sensitive to β-catenin inhibitor PKF118-310. Conclusion We have identified several overexpressed genes/pathways in thyroid cancer metastasis, making them attractive therapeutic targets. Given the complexity of metastasis involving multiple pathways (PD-L1, Mertk, IL6, COX-1/Tbxas1-TXA2), simultaneously targeting more than one of these pathways may be warranted to achieve better therapeutic effect for metastatic thyroid cancer.
Collapse
Affiliation(s)
- Minjing Zou
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Amal Qattan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Monther Al-Alwan
- Department of Cell Therapy and Immunobiology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hazem Ghebeh
- Department of Cell Therapy and Immunobiology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Naif Binjumah
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Latifa Al-Haj
- Department of Molecular Biomedicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Khalid S. A. Khabar
- Department of Molecular Biomedicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdulmohsen Altaweel
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Falah Almohanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdullah M. Assiri
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Abdelilah Aboussekhra
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Ali S. Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Yufei Shi
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Yikilmaz AS, Hernandez CMR. A Rare But Fatal Toxicity: Immune Checkpoint Inhibitor-Related Acquired Thrombotic Thrombocytopenic Purpura. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2025; 8:15-22. [PMID: 39811422 PMCID: PMC11728379 DOI: 10.36401/jipo-24-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 01/16/2025]
Abstract
Thrombotic thrombocytopenic purpura (TTP) is characterized by thrombotic microangiopathy resulting from decreased activation of the von Willebrand factor-cleaving protease (ADAMTS13). TTP can cause organ damage and is often fatal if the appropriate treatment is not started immediately. Although primary immune TTP is the most common form of TTP, secondary immune etiologies, including complications from immune checkpoint inhibitors (ICIs), have also been reported. ICIs are used as neoadjuvant and adjuvant therapy for metastatic and nonmetastatic solid tumors and hematologic cancers. ICIs stimulate the T-cell-mediated antitumor response, and the subsequent upregulation of the immune system can cause ICI-related adverse events (AEs). ICI-associated AEs may result in various hematological outcomes. Therefore, TTP, as a rare ICI-related AE, requires awareness. TTP has been mentioned as a rare ICIrAE in a few case reports. When using ICIs, the differential diagnosis of TTP should be considered if hemolytic anemia is accompanied by thrombocytopenia. Low ADAMTS13 activity can be used to diagnose TTP and support the need for plasma exchange. This review will assess the approach for ICI-related acquired TTP by scanning a limited number of reported case series in the literature. Low ADAMTS13 activity can be used to diagnose TTP and support the need for plasma exchange. Treatment in the cases that have been published includes combinations of rituximab and caplacizumab, corticosteroids, and plasma exchange. Furthermore, acquired TTP associated with ICI is encountered during the initial and subsequent cycles of ICI treatment. It is essential to detect ICI-related acquired TTP early, a highly fatal AE of ICIs, and to increase awareness of TTP, which will likely be encountered more frequently with the use of new ICI agents.
Collapse
|
15
|
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
|
16
|
Yao J, Lin X, Zhang X, Xie M, Ma X, Bao X, Song J, Liang Y, Wang Q, Xue X. Predictive biomarkers for immune checkpoint inhibitors therapy in lung cancer. Hum Vaccin Immunother 2024; 20:2406063. [PMID: 39415535 PMCID: PMC11487980 DOI: 10.1080/21645515.2024.2406063] [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: 04/23/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment mode of lung cancer, extending the survival time of patients unprecedentedly. Once patients respond to ICIs, the median duration of response is usually longer than that achieved with cytotoxic or targeted drugs. Unfortunately, there is still a large proportion of lung cancer patients do not respond to ICI. Effective biomarkers are crucial for identifying lung cancer patients who can benefit from them. The first predictive biomarker is programmed death-ligand 1 (PD-L1), but its predictive value is limited to specific populations. With the development of single-cell sequencing and spatial imaging technologies, as well as the use of deep learning and artificial intelligence, the identification of predictive biomarkers has been greatly expanded. In this review, we will dissect the biomarkers used to predict ICIs efficacy in lung cancer from the tumor-immune microenvironment and host perspectives, and describe cutting-edge technologies to further identify biomarkers.
Collapse
Affiliation(s)
- Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
17
|
Bravaccini S, Boldrin E, Gurioli G, Tedaldi G, Piano MA, Canale M, Curtarello M, Ulivi P, Pilati P. The use of platelets as a clinical tool in oncology: opportunities and challenges. Cancer Lett 2024; 607:217044. [PMID: 38876385 DOI: 10.1016/j.canlet.2024.217044] [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: 02/16/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
Platelets are small circulating anucleated cells mainly involved in thrombosis and hemostasis processes. Moreover, platelets play an active role in tumorigenesis and cancer progression, stimulating angiogenesis and vascular remodelling, and protecting circulating cancer cells from shear forces and immune surveillance. Several reports indicate that platelet number in the blood circulation of cancer patients is associated with prognosis and response to treatment. However, the mechanisms of platelets "education" by cancer cells and the crosstalk between platelets and tumor are still unclear, and the role of "tumor educated platelets" (TEPs) is achieving growing interest in cancer research. TEPs are a biological source of cancer-derived biomarkers, especially RNAs that are protected by platelets membrane from circulating RNases, and could serve as a non-invasive tool for tumor detection, molecular profiling and evolution during therapy in clinical practice. Moreover, short platelet lifespan offers the possibility to get a snapshot assessment of cancer molecular profile, providing a real-time tool. We review and discuss the potential and the clinical utility, in terms of cancer diagnosis and monitoring, of platelet count together with other morphological parameters and of the more recent and innovative TEP profiling.
Collapse
Affiliation(s)
- Sara Bravaccini
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Elisa Boldrin
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Giorgia Gurioli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Gianluca Tedaldi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Maria Assunta Piano
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Matteo Canale
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Matteo Curtarello
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padua, Italy.
| | - Paola Ulivi
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", via P. Maroncelli 40, 47014, Meldola, Italy.
| | - Pierluigi Pilati
- Surgical Oncology of Digestive Tract Unit, Veneto Institute of Oncology IOV-IRCCS, 35128, Padova, Italy.
| |
Collapse
|
18
|
Dou X, Li K, Lu J. Obinutuzumab-induced acute thrombocytopenia: a case report and literature review. Front Oncol 2024; 14:1509567. [PMID: 39741978 PMCID: PMC11685111 DOI: 10.3389/fonc.2024.1509567] [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: 10/11/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Background Obinutuzumab, a humanized type II anti-CD20 monoclonal antibody, is widely used in the treatment of B-cell lymphomas. Thrombocytopenia typically occurs 1 to 2 weeks after administration. In rare cases, obinutuzumab can induce severe acute thrombocytopenia within days of infusion, a condition known as "obinutuzumab-induced acute thrombocytopenia (OIAT)." Rituximab, a chimeric type I anti-CD20 monoclonal antibody, is also known to cause "rituximab-induced acute thrombocytopenia (RIAT)." This report presents a case of OIAT, with subsequent treatment switched to rituximab, which did not result in thrombocytopenia recurrence. Case Presentation A 38-year-old female patient with a 2-year history of lymphadenopathy was diagnosed with follicular lymphoma (Grade I-II). She was treated with obinutuzumab combined with bendamustine. Following the first administration of obinutuzumab, her platelet count dropped to 37×10⁹/L within 2 days and further declined to 27×10⁹/L on the fourth day without bleeding symptoms. The platelet count recovered by day 8. After a second obinutuzumab infusion, the platelet count again dropped to 15×10⁹/L within 1 day. Platelet transfusion was effective, and the count eventually recovered to 95×10⁹/L by day 29. No further acute thrombocytopenia occurred after switching to rituximab. Conclusion OIAT is a rare but serious adverse effect of obinutuzumab. This case highlights the importance of early recognition and monitoring of platelet counts in patients receiving obinutuzumab. The findings in our case, along with those in the literature, suggest that switching to rituximab or extending the interval before obinutuzumab re-administration can reduce the risk of recurrent thrombocytopenia. Further research is needed to elucidate the underlying mechanisms and establish treatment guidelines for OIAT.
Collapse
Affiliation(s)
- Xuelin Dou
- Department of Hematology, Peking University People’s Hospital, National Clinical Research Center for Hematologic Diseases, Peking University Institute of Hematology, Beijing, China
| | - Kongyang Li
- Department of Hematology, Peking University People’s Hospital, National Clinical Research Center for Hematologic Diseases, Peking University Institute of Hematology, Beijing, China
- Department of Hematology, People’s Hospital of Yangjiang, Yangjiang, China
| | - Jin Lu
- Department of Hematology, Peking University People’s Hospital, National Clinical Research Center for Hematologic Diseases, Peking University Institute of Hematology, Beijing, China
| |
Collapse
|
19
|
Zhu J, Zhang W, Wang Z, Wang Y, Li J, Wang Y, Xu F, Chen Y. Mass-tagged self-assembled nanoprobe reveals the transport of PD-L1 from cancer cells to tumor-educated platelets. Anal Chim Acta 2024; 1331:343312. [PMID: 39532409 DOI: 10.1016/j.aca.2024.343312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The expression level of immune checkpoint proteins detected by tissue biopsy is currently used as a predictive biomarker for immune checkpoint blockade (ICB) therapy. However, tissue biopsy is susceptible to invasive sample collection procedures, significant sampling heterogeneity, and the difficulty of repeated sampling. Therefore, liquid biopsy of blood samples is becoming an alternative choice for immune checkpoint protein detection. Among various vesicles in blood, platelets can obtain cancer information to form a specific group called tumor-educated platelets (TEPs). The platelet-derived proteins in TEPs may have a predictive potential in ICB therapy. RESULTS In this study, a photo-cleavable mass-tagged self-assembled (SAMT) nanoprobe with signal amplification was developed for the quantitative detection of PD-L1. The SAMT probe was assembled by photo-cleavable mass tags, PD-L1 aptamer, and amphiphilic polymer. After binding with PD-L1 on the platelet, the probe can release mass tags with UV light exposure. The amount of the mass tag, representing that of PD-L1, was subsequently determined by mass spectrometry. The assay sensitivity can be greatly improved by up to four orders of magnitude, achieving a detection limit of 10 fM. This assay was subsequently applied to cancer cells and platelet samples from non-small cell lung cancer (NSCLC) patients. The patients with higher tumor stages, higher degrees of lymph node invasion, and better ICB response had higher PD-L1 levels on platelets. Further investigation revealed that PD-L1 on the platelets was transported from cancer cells, providing evidence for the existence of TEPs. SIGNIFICANCE The SAMT probe can amplify the signal of the target molecule into that of multiple mass tags, achieving ultrasensitive ICB protein quantitative detection in platelets. Moreover, the employed SAMT assay not only revealed PD-L1 transport from cancer cells to platelets but also confirmed the presence of TEPs.
Collapse
Affiliation(s)
- Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Wenjun Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhongcheng Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiapu Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yunjing Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feifei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing, 211166, China; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing, 211166, China.
| |
Collapse
|
20
|
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
|
21
|
Chen L, Rao W, Chen Y, Xie J. In vitro induction of anti‑lung cancer immune response by the A549 lung cancer stem cell lysate‑sensitized dendritic cell vaccine. Oncol Lett 2024; 28:550. [PMID: 39328277 PMCID: PMC11425031 DOI: 10.3892/ol.2024.14683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Lung adenocarcinoma is one of the most fatal types of cancer worldwide, with non-small cell lung cancer being the most common subtype. Therefore, there is need for improved treatment approaches. Tumor growth results from the proliferation of a very small number of tumor stem cells, giving rise to the theory of cancer stem cells (CSCs). Lung CSCs are associated with lung cancer development, and although chemotherapy drugs can inhibit the proliferation of lung cancer cells, they have difficulty acting on lung CSCs. Even if the tumor appears to have disappeared after chemotherapy, the presence of a small number of residual tumor stem cells can lead to cancer recurrence and metastasis. Hence, targeting and eliminating lung CSCs is of significant therapeutic importance. In this study, we cultured A549 cells in sphere-forming conditions using B27, EGF, and bFGF, isolated peripheral blood mononuclear cells (PBMCs), and induced and characterized dendritic cells (DCs). We also isolated and expanded T lymphocytes. DC vaccines were prepared using A549 stem cell lysate or A549 cell lysate for sensitization and compared with non-sensitized DC vaccines. The content of IFN-γ in the supernatant of cultures with vaccines and T cells was measured by ELISA. The cytotoxic effects of the vaccines on A549 cells and stem cells were assessed using the Cytotox96 assay, and the impact of the vaccines on A549 cell migration and apoptosis was evaluated using Transwell assays and flow cytometry. DC vaccines sensitized with human lung CSC lysates induced significant in vitro cytotoxic effects on A549 lung cancer cells and CSCs by T lymphocytes, while not producing immune cytotoxic effects on human airway epithelial cells. Moreover, the immune-killing effect induced by DC vaccines sensitized with lung CSC lysates was superior to that of DC vaccines sensitized with lung cancer cells.
Collapse
Affiliation(s)
- Letian Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Wei Rao
- Department of Urology, Yingtan People's Hospital, Yingtan, Jiangxi 335000, P.R. China
| | - Yujuan Chen
- Department of Pulmonary and Critical Care Medicine, Gaoan People's Hospital, Yichun, Jiangxi 336000, P.R. China
| | - Junping Xie
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
22
|
Ghosh DD, McDonald H, Dutta R, Krishnan K, Thilakan J, Paul MK, Arya N, Rao M, Rangnekar VM. Prognostic Indicators for Precision Treatment of Non-Small Cell Lung Carcinoma. Cells 2024; 13:1785. [PMID: 39513892 PMCID: PMC11545304 DOI: 10.3390/cells13211785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) has established predictive biomarkers that enable decisions on treatment regimens for many patients. However, resistance to therapy is widespread. It is therefore essential to have a panel of molecular biomarkers that may help overcome therapy resistance and prevent adverse effects of treatment. We performed in silico analysis of NSCLC prognostic indicators, separately for adenocarcinomas and squamous carcinomas, by using The Cancer Genome Atlas (TCGA) and non-TCGA data sources in cBioPortal as well as UALCAN. This review describes lung cancer biology, elaborating on the key genetic alterations and specific genes responsible for resistance to conventional treatments. Importantly, we examined the mechanisms associated with resistance to immune checkpoint inhibitors. Our analysis indicated that a robust prognostic biomarker was lacking for NSCLC, especially for squamous cell carcinomas. In this work, our screening uncovered previously unidentified prognostic gene expression indicators, namely, MYO1E, FAM83 homologs, and DKK1 for adenocarcinoma, and FGA and TRIB1 for squamous cell carcinoma. It was further observed that overexpression of these genes was associated with poor prognosis. Additionally, FAM83 homolog and TRIB1 unexpectedly harbored copy number amplifications. In conclusion, this study elucidated novel prognostic indicators for NSCLC that may serve as targets to overcome therapy resistance toward improved patient outcomes.
Collapse
Affiliation(s)
- Damayanti Das Ghosh
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata 700063, West Bengal, India; (D.D.G.); (R.D.)
- School of Health Sciences and Translational Research, Sister Nivedita University, Newtown, Kolkata 700156, West Bengal, India
| | - Hannah McDonald
- Department of Surgery, University of Kentucky, Lexington, KY 40536, USA;
| | - Rajeswari Dutta
- Basic and Translational Research Division, Saroj Gupta Cancer Centre and Research Institute, Mahatma Gandhi Road, Kolkata 700063, West Bengal, India; (D.D.G.); (R.D.)
| | - Keerthana Krishnan
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Jaya Thilakan
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India;
- Department of Genetics, UTD, Barkatullah University Bhopal, Bhopal 462026, Madhya Pradesh, India
| | - Manash K. Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Neha Arya
- Department of Translational Medicine, All India Institute of Medical Sciences Bhopal, Bhopal 462020, Madhya Pradesh, India;
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India;
| | - Vivek M. Rangnekar
- Markey Cancer Center and Department of Radiation Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
23
|
Zhang M, Li YP, He N, Dang SS. Platelets in liver cancer. Shijie Huaren Xiaohua Zazhi 2024; 32:735-741. [DOI: 10.11569/wcjd.v32.i10.735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Affiliation(s)
- Meng Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 71004, Shaanxi Province, China
| | - Ya-Ping Li
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 71004, Shaanxi Province, China
| | - Na He
- Department of Gastroenterology, First Affiliated Hospital of Xi'an Medical College, Xi'an 710077, Shaanxi Province, China
| | - Shuang-Suo Dang
- Department of Infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 71004, Shaanxi Province, China
| |
Collapse
|
24
|
Wang Q, Wu T, Zhao R, Li Y, Chen X, Shen S, Zhang X. Development and Validation of a Prediction Model for Thyroid Dysfunction in Patients During Immunotherapy. Endocr Pract 2024; 30:943-950. [PMID: 39004306 DOI: 10.1016/j.eprac.2024.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/24/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
OBJECTIVE This study was designed to develop and validate a predictive model for assessing the risk of thyroid toxicity following treatment with immune checkpoint inhibitors. METHODS A retrospective analysis was conducted on a cohort of 586 patients diagnosed with malignant tumors who received programmed cell death 1 (PD-1)/programmed death-ligand 1 (PD-L1) inhibitors. The patients were randomly divided into training and validation cohorts in a 7:3 ratio. Logistic regression analyses were performed on the training set to identify risk factors of thyroid dysfunction, and a nomogram was developed based on these findings. Internal validation was performed using K-fold cross-validation on the validation set. The performance of the nomogram was assessed in terms of discrimination and calibration. Additionally, decision curve analysis was utilized to demonstrate the decision efficiency of the model. RESULTS Our clinical prediction model consisted of 4 independent predictors of thyroid immune-related adverse events, namely baseline thyrotropin (TSH, OR = 1.427, 95%CI:1.163-1.876), baseline thyroglobulin antibody (TgAb, OR = 1.105, 95%CI:1.035-1.180), baseline thyroid peroxidase antibody (TPOAb, OR = 1.172, 95%CI:1.110-1.237), and baseline platelet count (platelet, OR = 1.004, 95%CI:1.000-1.007). The developed nomogram achieved excellent discrimination with an area under the curve of 0.863 (95%CI: 0.817-0.909) and 0.885 (95%CI: 0.827-0.944) in the training and internal validation cohorts respectively. Calibration curves exhibited a good fit, and the decision curve indicated favorable clinical benefits. CONCLUSION The proposed nomogram serves as an effective and intuitive tool for predicting the risk of thyroid immune-related adverse events, facilitating clinicians making individualized decisions based on patient-specific information.
Collapse
Affiliation(s)
- Qian Wang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Tingting Wu
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Ru Zhao
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China; Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuanqin Li
- Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China; Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuetao Chen
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China
| | - Shanmei Shen
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| | - Xiaowen Zhang
- Department of Endocrinology, Endocrine and Metabolic Disease Medical Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China; Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing, China.
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Chen C, Zhang J, Liu X, Zhuang Q, Lu H, Hou J. A platelet-related signature for predicting the prognosis and immunotherapy benefit in bladder cancer based on machine learning combinations. Transl Androl Urol 2024; 13:1472-1485. [PMID: 39280688 PMCID: PMC11399026 DOI: 10.21037/tau-24-80] [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: 02/09/2024] [Accepted: 06/26/2024] [Indexed: 09/18/2024] Open
Abstract
Background Bladder cancer carries a large societal burden, with over 570,000 newly diagnosed cases and 210,000 deaths globally each year. Platelets play vital functions in tumor progression and therapy benefits. We aimed to construct a platelet-related signature (PRS) for the clinical outcome of bladder cancer cases. Methods Ten machine learning techniques were used in the integrative operations to build PRS using the datasets from The Cancer Genome Atlas (TCGA), gene series expression (GSE)13507, GSE31684, GSE32894 and GSE48276. A number of immunotherapy datasets and prediction scores, including GSE91061, GSE78220, and IMvigor210, were utilized to assess how well the PRS predicted the benefit of immunotherapy. Vitro experiment was performed to verify the role of α1C-tubulin (TUBA1C) in bladder cancer. Results Enet (alpha =0.4) algorithm-based PRS had the highest average C-index of 0.73 and it was suggested as the optimal PRS. PRS acted as an independent risk factor for bladder cancer and patients with high PRS score portended a worse overall survival rate, with the area under the curve of 1-, 3- and 5-year operating characteristic curve being 0.754, 0.779 and 0.806 in TCGA dataset. A higher level of immune-activated cells, cytolytic function and T cell co-stimulation was found in the low PRS score group. Low PRS score demonstrated a higher tumor mutation burden score and programmed cell death protein 1 & cytotoxic T-lymphocyte associated protein 4 immunophenoscore, lower tumor immune dysfunction and exclusion score, intratumor heterogeneity score and immune escape score in bladder cancer, suggesting the PRS as an indicator for predicting immunotherapy benefits. Vitro experiment showed that TUBA1C was upregulated in bladder cancer and knockdown of TUBA1C obviously suppressed tumor cell proliferation. Conclusions The present study developed an ideal PRS for bladder cancer, which may be used as a predictor of prognosis, a risk classification system, and a therapy guide.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Zhang
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoshuang Liu
- Department of General Surgery, Shuguang Hospital, Shanghai University of traditional Chinese Medicine, Shanghai, China
| | - Qianfeng Zhuang
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Lu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianquan Hou
- Department of Urology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, China
| |
Collapse
|
28
|
Schaubaecher JB, Smiljanov B, Haring F, Steiger K, Wu Z, Ugurluoglu A, Luft J, Ballke S, Mahameed S, Schneewind V, Hildinger J, Canis M, Mittmann LA, Braun C, Zuchtriegel G, Kaiser R, Nicolai L, Mack M, Weichert W, Lauber K, Uhl B, Reichel CA. Procoagulant platelets promote immune evasion in triple-negative breast cancer. Blood 2024; 144:216-226. [PMID: 38648571 DOI: 10.1182/blood.2023022928] [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: 10/16/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) is an aggressive tumor entity in which immune checkpoint (IC) molecules are primarily synthesized in the tumor environment. Here, we report that procoagulant platelets bear large amounts of such immunomodulatory factors and that the presence of these cellular blood components in TNBC relates to protumorigenic immune-cell activity and impaired survival. Mechanistically, tumor-released nucleic acids attract platelets to the aberrant tumor microvasculature, where they undergo procoagulant activation, thus delivering specific stimulatory and inhibitory IC molecules. This concomitantly promotes protumorigenic myeloid leukocyte responses and compromises antitumorigenic lymphocyte activity, ultimately supporting tumor growth. Interference with platelet-leukocyte interactions prevented immune cell misguidance and suppressed tumor progression, nearly as effective as systemic IC inhibition. Hence, our data uncover a self-sustaining mechanism of TNBC by using platelets to misdirect immune-cell responses. Targeting this irregular multicellular interplay may represent a novel immunotherapeutic strategy for TNBC without the adverse effects of systemic IC inhibition.
Collapse
Affiliation(s)
- Johanna B Schaubaecher
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Bojan Smiljanov
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Florian Haring
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Katja Steiger
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Zhengquan Wu
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Anais Ugurluoglu
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Joshua Luft
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Simone Ballke
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Shaan Mahameed
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Vera Schneewind
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Jonas Hildinger
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Comprehensive Cancer Center, Munich Ludwig-Maximilians-Universität, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Laura A Mittmann
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Constanze Braun
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Rainer Kaiser
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Berlin, Germany
| | - Leo Nicolai
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Berlin, Germany
| | - Matthias Mack
- Department of Nephrology, University of Regensburg, Regensburg, Germany
| | - Wilko Weichert
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Bernd Uhl
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Christoph A Reichel
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Comprehensive Cancer Center, Munich Ludwig-Maximilians-Universität, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| |
Collapse
|
29
|
Di Mauro F, Arbore G. Spatial Dissection of the Immune Landscape of Solid Tumors to Advance Precision Medicine. Cancer Immunol Res 2024; 12:800-813. [PMID: 38657223 PMCID: PMC11217735 DOI: 10.1158/2326-6066.cir-23-0699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 01/12/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Chemotherapeutics, radiation, targeted therapeutics, and immunotherapeutics each demonstrate clinical benefits for a small subset of patients with solid malignancies. Immune cells infiltrating the tumor and the surrounding stroma play a critical role in shaping cancer progression and modulating therapy response. They do this by interacting with the other cellular and molecular components of the tumor microenvironment. Spatial multi-omics technologies are rapidly evolving. Currently, such technologies allow high-throughput RNA and protein profiling and retain geographical information about the tumor microenvironment cellular architecture and the functional phenotype of tumor, immune, and stromal cells. An in-depth spatial characterization of the heterogeneous tumor immune landscape can improve not only the prognosis but also the prediction of therapy response, directing cancer patients to more tailored and efficacious treatments. This review highlights recent advancements in spatial transcriptomics and proteomics profiling technologies and the ways these technologies are being applied for the dissection of the immune cell composition in solid malignancies in order to further both basic research in oncology and the implementation of precision treatments in the clinic.
Collapse
Affiliation(s)
- Francesco Di Mauro
- Vita-Salute San Raffaele University, Milan, Italy.
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Giuseppina Arbore
- Vita-Salute San Raffaele University, Milan, Italy.
- Experimental Immunology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
30
|
Tuerhong N, Yang Y, Wang C, Huang P, Li Q. Interactions between platelets and the cancer immune microenvironment. Crit Rev Oncol Hematol 2024; 199:104380. [PMID: 38718939 DOI: 10.1016/j.critrevonc.2024.104380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/16/2024] Open
Abstract
Cancer is a leading cause of death in both China and developed countries due to its high incidence and low cure rate. Immune function is closely linked to the development and progression of tumors. Platelets, which are primarily known for their role in hemostasis, also play a crucial part in the spread and progression of tumors through their interaction with the immune microenvironment. The impact of platelets on tumor growth and metastasis depends on the type of cancer and treatment method used. This article provides an overview of the relationship between platelets and the immune microenvironment, highlighting how platelets can either protect or harm the immune response and cancer immune escape. We also explore the potential of available platelet-targeting strategies for tumor immunotherapy, as well as the promise of new platelet-targeted tumor therapy methods through further research.
Collapse
Affiliation(s)
- Nuerye Tuerhong
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Yang Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Chenyu Wang
- The Second Clinical Medical College, Lanzhou university, No. 222 South Tianshui Road, Gansu, China
| | - Peng Huang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China; West China Biomedical Big Data Center, Sichuan University, No. 37, GuoXue Xiang Chengdu, Sichuan, China.
| |
Collapse
|
31
|
Wang X, Bai L, Kong L, Guo Z. Advances in circulating tumor cells for early detection, prognosis and metastasis reduction in lung cancer. Front Oncol 2024; 14:1411731. [PMID: 38974237 PMCID: PMC11224453 DOI: 10.3389/fonc.2024.1411731] [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: 04/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Globally, lung cancer stands as the leading type of cancer in terms of incidence and is the major source of mortality attributed to cancer. We have outlined the molecular biomarkers for lung cancer that are available clinically. Circulating tumor cells (CTCs) spread from the original location, circulate in the bloodstream, extravasate, and metastasize, forming secondary tumors by invading and establishing a favorable environment. CTC analysis is considered a common liquid biopsy method for lung cancer. We have enumerated both in vivo and ex vivo techniques for CTC separation and enrichment, examined the advantages and limitations of these methods, and also discussed the detection of CTCs in other bodily fluids. We have evaluated the value of CTCs, as well as CTCs in conjunction with other biomarkers, for their utility in the early detection and prognostic assessment of patients with lung cancer. CTCs engage with diverse cells of the metastatic process, interfering with the interaction between CTCs and various cells in metastasis, potentially halting metastasis and enhancing patient prognosis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Lu Bai
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Linghui Kong
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Zhijuan Guo
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
32
|
Shi Q, Xu J, Chen C, Hu X, Wang B, Zeng F, Ren T, Huang Y, Guo W, Tang X, Ji T. Direct contact between tumor cells and platelets initiates a FAK-dependent F3/TGF-β positive feedback loop that promotes tumor progression and EMT in osteosarcoma. Cancer Lett 2024; 591:216902. [PMID: 38641310 DOI: 10.1016/j.canlet.2024.216902] [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: 02/10/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Platelets have received growing attention for their roles in hematogenous tumor metastasis. However, the tumor-platelet interaction in osteosarcoma (OS) remains poorly understood. Here, using platelet-specific focal adhesion kinase (FAK)-deficient mice, we uncover a FAK-dependent F3/TGF-β positive feedback loop in OS. Disruption of the feedback loop by inhibition of F3, TGF-β, or FAK significantly suppresses OS progression. We demonstrate that OS F3 initiated the feedback loop by increasing platelet TGF-β secretion, and platelet-derived TGF-β promoted OS F3 expression in turn and modulated OS EMT process. Immunofluorescence results indicate platelet infiltration in OS niche and we verified it was mediated by platelet FAK. In addition, platelet FAK was proved to mediate platelet adhesion to OS cells, which was vital for the initiation of F3/TGF-β feedback loop. Collectively, these findings provide a rationale for novel therapeutic strategies targeting tumor-platelet interplay in metastatic OS.
Collapse
Affiliation(s)
- Qianyu Shi
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Chenglong Chen
- Department of Orthopedics, Beijing Jishuitan Hospital, Beijing, China
| | - Xueyu Hu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Fanwei Zeng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Yi Huang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Tao Ji
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| |
Collapse
|
33
|
Moaveni AK, Amiri M, Shademan B, Farhadi A, Behroozi J, Nourazarian A. Advances and challenges in gene therapy strategies for pediatric cancer: a comprehensive update. Front Mol Biosci 2024; 11:1382190. [PMID: 38836106 PMCID: PMC11149429 DOI: 10.3389/fmolb.2024.1382190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 06/06/2024] Open
Abstract
Pediatric cancers represent a tragic but also promising area for gene therapy. Although conventional treatments have improved survival rates, there is still a need for targeted and less toxic interventions. This article critically analyzes recent advances in gene therapy for pediatric malignancies and discusses the challenges that remain. We explore the innovative vectors and delivery systems that have emerged, such as adeno-associated viruses and non-viral platforms, which show promise in addressing the unique pathophysiology of pediatric tumors. Specifically, we examine the field of chimeric antigen receptor (CAR) T-cell therapies and their adaptation for solid tumors, which historically have been more challenging to treat than hematologic malignancies. We also discuss the genetic and epigenetic complexities inherent to pediatric cancers, such as tumor heterogeneity and the dynamic tumor microenvironment, which pose significant hurdles for gene therapy. Ethical considerations specific to pediatric populations, including consent and long-term follow-up, are also analyzed. Additionally, we scrutinize the translation of research from preclinical models that often fail to mimic pediatric cancer biology to the regulatory landscapes that can either support or hinder innovation. In summary, this article provides an up-to-date overview of gene therapy in pediatric oncology, highlighting both the rapid scientific progress and the substantial obstacles that need to be addressed. Through this lens, we propose a roadmap for future research that prioritizes the safety, efficacy, and complex ethical considerations involved in treating pediatric patients. Our ultimate goal is to move from incremental advancements to transformative therapies.
Collapse
Affiliation(s)
- Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Farhadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Javad Behroozi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
34
|
Anguera G, Mulet M, Zamora C, Osuna-Gómez R, Barba A, Sullivan I, Serra-López J, Cantó E, Vidal S, Majem M. Potential Role of Circulating PD-L1 + Leukocytes as a Predictor of Response to Anti-PD-(L)1 Therapy in NSCLC Patients. Biomedicines 2024; 12:958. [PMID: 38790920 PMCID: PMC11117542 DOI: 10.3390/biomedicines12050958] [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: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
PD-(L)1 inhibitors are part of the treatment strategy for non-small cell lung cancer (NSCLC) although its efficacy is limited to certain patients. Our study aimed to identify patients who might benefit from anti-PD-(L)1 inhibitors by analyzing the PD-L1 expression on circulating leukocytes and its evolution during treatment. One hundred thirteen NSCLC patients, according to their radiological response after 10-12 weeks of treatment, were classified into responders, stable, and progressive disease. Percentages of circulating PD-L1+ leukocytes, PD-L1+ platelets (PLTs), and leukocyte-PLT complexes were assessed using flow cytometry, and plasma concentrations of soluble immunomodulatory factors were quantified by ELISA. Responders exhibited significantly higher pre-treatment percentages of PD-L1+ neutrophils, PD-L1+ CD14+ cells, and PD-L1+ PLTs than progressors. The percentages of these populations decreased in responders post-treatment, contrasting with stables and progressors. PLTs notably contributed to PD-L1 expression in CD14+ cells and neutrophils. Plasma cytokine analysis revealed baseline differences only in IL-17 concentration among groups, whereas network analyses highlighted distinct association patterns between plasma molecules and PD-L1+ leukocytes after 10-12 weeks of treatment. Our findings suggest that pre-treatment assessment of circulating PD-L1+ neutrophils, PD-L1+ CD14+ cells, and PD-L1+ PLTs may be helpful in identifying NSCLC patients who are potential candidates for anti-PD-(L)1 therapy.
Collapse
Affiliation(s)
- Georgia Anguera
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Maria Mulet
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Carlos Zamora
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Rubén Osuna-Gómez
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Andrés Barba
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Ivana Sullivan
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Jorgina Serra-López
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| | - Elisabet Cantó
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain; (C.Z.); (R.O.-G.); (E.C.); (S.V.)
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (G.A.); (A.B.); (I.S.); (J.S.-L.); (M.M.)
| |
Collapse
|
35
|
Wang ZY, Xu B, Wang LN, Zhu XD, Huang C, Shen YH, Li H, Li ML, Zhou J, Fan J, Sun HC. Platelet-to-lymphocyte ratio predicts tumor response and survival of patients with hepatocellular carcinoma undergoing immunotherapies. Int Immunopharmacol 2024; 131:111863. [PMID: 38492340 DOI: 10.1016/j.intimp.2024.111863] [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: 12/22/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Lymphocyte-related factors were associated with survival outcome of different types of cancers. Nevertheless, the association between lymphocytes-related factors and tumor response of immunotherapy remains unclear. METHODS This is a retrospective study. Eligible participants included patients with unresectable or advanced hepatocellular carcinoma (HCC) who underwent immunotherapy as their first-line treatment. Radiological assessment of tumor response adhered to RECIST 1.1 and HCC-specific modified RECIST (mRECIST) criteria. Univariate and multivariate logistic analyses were employed to analyze clinical factors associated with tumor response. Kaplan-Meier survivial analysis were employed to compare progression-free survival (PFS) and overall survival (OS) across different clinical factors. Furthermore, patients who received treatment with either a combination of bevacizumab and anti-PD-1(L1) antibody (Beva group) or tyrosine-kinase inhibitor (TKI) and anti-PD-1 antibody (TKI group) were examined to explore the relation between clinical factors and tumor response. RESULTS A total of 208 patients were enrolled in this study. The median PFS and OS were 9.84 months and 24.44 months,respectively. An independent factor associated with a more favorable tumor response to immunotherapy was identified when PLR<100. Patients with PLR<100 had longer PFS than other patients, while OS showed no significant difference. Further analysis revealed that PLR exhibited superior prognostic value in patients of the Beva group as compared to those in the TKI group. CONCLUSIONS There exisits an association between PLR and tumor response as well as survival outcomes in patients receiving immunotherapy, particularly those treated with the combination of bevacizumab and anti-PD-1.
Collapse
Affiliation(s)
- Zi-Yi Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lu-Na Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mei-Ling Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
36
|
Xue J, Deng J, Qin H, Yan S, Zhao Z, Qin L, Liu J, Wang H. The interaction of platelet-related factors with tumor cells promotes tumor metastasis. J Transl Med 2024; 22:371. [PMID: 38637802 PMCID: PMC11025228 DOI: 10.1186/s12967-024-05126-6] [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: 01/16/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Platelets not only participate in thrombosis and hemostasis but also interact with tumor cells and protect them from mechanical damage caused by hemodynamic shear stress and natural killer cell lysis, thereby promoting their colonization and metastasis to distant organs. Platelets can affect the tumor microenvironment via interactions between platelet-related factors and tumor cells. Metastasis is a key event in cancer-related death and is associated with platelet-related factors in lung, breast, and colorectal cancers. Although the factors that promote platelet expression vary slightly in terms of their type and mode of action, they all contribute to the overall process. Recognizing the correlation and mechanisms between these factors is crucial for studying the colonization of distant target organs and developing targeted therapies for these three types of tumors. This paper reviews studies on major platelet-related factors closely associated with metastasis in lung, breast, and colorectal cancers.
Collapse
Affiliation(s)
- Jie Xue
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Jianzhao Deng
- Clinical Laboratory, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Hongwei Qin
- Department of Blood Transfusion, The Central Hospital of Qingdao Jiaozhou, 99 Yunxi River South Road, Qingdao, 266300, Shandong, China
| | - Songxia Yan
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Zhen Zhao
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Lifeng Qin
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Jiao Liu
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, 266000, Shandong, China.
| |
Collapse
|
37
|
Huelsboemer L, Knoedler L, Kochen A, Yu CT, Hosseini H, Hollmann KS, Choi AE, Stögner VA, Knoedler S, Hsia HC, Pomahac B, Kauke-Navarro M. Cellular therapeutics and immunotherapies in wound healing - on the pulse of time? Mil Med Res 2024; 11:23. [PMID: 38637905 PMCID: PMC11025282 DOI: 10.1186/s40779-024-00528-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Chronic, non-healing wounds represent a significant challenge for healthcare systems worldwide, often requiring significant human and financial resources. Chronic wounds arise from the complex interplay of underlying comorbidities, such as diabetes or vascular diseases, lifestyle factors, and genetic risk profiles which may predispose extremities to local ischemia. Injuries are further exacerbated by bacterial colonization and the formation of biofilms. Infection, consequently, perpetuates a chronic inflammatory microenvironment, preventing the progression and completion of normal wound healing. The current standard of care (SOC) for chronic wounds involves surgical debridement along with localized wound irrigation, which requires inpatient care under general anesthesia. This could be followed by, if necessary, defect coverage via a reconstructive ladder utilizing wound debridement along with skin graft, local, or free flap techniques once the wound conditions are stabilized and adequate blood supply is restored. To promote physiological wound healing, a variety of approaches have been subjected to translational research. Beyond conventional wound healing drugs and devices that currently supplement treatments, cellular and immunotherapies have emerged as promising therapeutics that can behave as tailored therapies with cell- or molecule-specific wound healing properties. However, in contrast to the clinical omnipresence of chronic wound healing disorders, there remains a shortage of studies condensing the current body of evidence on cellular therapies and immunotherapies for chronic wounds. This review provides a comprehensive exploration of current therapies, experimental approaches, and translational studies, offering insights into their efficacy and limitations. Ultimately, we hope this line of research may serve as an evidence-based foundation to guide further experimental and translational approaches and optimize patient care long-term.
Collapse
Affiliation(s)
- Lioba Huelsboemer
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Leonard Knoedler
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- School of Medicine, University of Regensburg, 93040, Regensburg, Germany
| | - Alejandro Kochen
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- Regenerative Wound Healing Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Catherine T Yu
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Helia Hosseini
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Katharina S Hollmann
- School of Medicine, University of Wuerzburg, 97070, Würzburg, Germany
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Ashley E Choi
- California University of Science and Medicine, Colton, CA, 92324, USA
| | - Viola A Stögner
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Samuel Knoedler
- School of Medicine, University of Regensburg, 93040, Regensburg, Germany
| | - Henry C Hsia
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
- Regenerative Wound Healing Center, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Bohdan Pomahac
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA
| | - Martin Kauke-Navarro
- Division of Reconstructive and Plastic Surgery, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
38
|
Xu J, Zhao Y, Chen Z, Wei L. Clinical Application of Different Liquid Biopsy Components in Hepatocellular Carcinoma. J Pers Med 2024; 14:420. [PMID: 38673047 PMCID: PMC11051574 DOI: 10.3390/jpm14040420] [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: 02/23/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, usually occurring in the background of chronic liver disease. HCC lethality rate is in the third highest place in the world. Patients with HCC have concealed early symptoms and possess a high-level of heterogeneity. Once diagnosed, most of the tumors are in advanced stages and have a poor prognosis. The sensitivity and specificity of existing detection modalities and protocols are suboptimal. HCC calls for more sophisticated and individualized therapeutic regimens. Liquid biopsy is non-invasive, repeatable, unaffected by location, and can be monitored dynamically. It has emerged as a useable aid in achieving precision malignant tumor treatment. Circulating tumor cells (CTCs), circulating nucleic acids, exosomes and tumor-educated platelets are the commonest components of a liquid biopsy. It possesses the theoretical ability to conquer the high heterogeneity and the difficulty of early detection for HCC patients. In this review, we summarize the common enrichment techniques and the clinical applications in HCC for different liquid biopsy components. Tumor recurrence after HCC-related liver transplantation is more insidious and difficult to treat. The clinical use of liquid biopsy in HCC-related liver transplantation is also summarized in this review.
Collapse
Affiliation(s)
| | | | | | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China; (J.X.); (Y.Z.); (Z.C.)
| |
Collapse
|
39
|
Abstract
Alongside their conventional roles in thrombosis and hemostasis, platelets have long been associated with nonhemostatic pathologies, including tumor cell metastasis. Numerous mechanistic studies have since demonstrated that the direct binding of platelets to intravascular tumor cells promotes key hallmarks of metastasis, including survival in circulation and tumor cell arrest at secondary sites. However, platelets also interact with nonmalignant cells that make up the stromal and immune compartments within both primary and metastatic tumors. This review will first provide a brief historical perspective on platelet contributions to metastatic disease before discussing the emerging roles that platelets play in creating microenvironments that likely support successful tumor cell metastasis.
Collapse
Affiliation(s)
- Harvey G. Roweth
- Hematology Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
40
|
Chen M, Wang S. Preclinical development and clinical studies of targeted JAK/STAT combined Anti-PD-1/PD-L1 therapy. Int Immunopharmacol 2024; 130:111717. [PMID: 38387193 DOI: 10.1016/j.intimp.2024.111717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
Programmed cell death protein 1 (PD-1) binds to its ligand to help tumours evade the immune system and promote tumour progression. Although anti-PD-1/PD-L1 therapies show powerful effects in some patients, most patients are unable to benefit from this treatment due to treatment resistance. Therefore, it is important to overcome tumour resistance to PD-1/PD-L1 blockade. There is substantial evidence suggesting that the JAK/STAT signalling pathway plays a significant role in PD-1/PD-L1 expression and anti-PD-1/PD-L1 treatment. Herein, we describe the effects of the JAK/STAT signalling pathway on PD-1/PD-L1. Subsequently, the relationship between molecular mutations in the JAK/STAT signalling pathway and immune resistance was analysed. Finally, the latest advancements in drugs targeting the JAK/STAT pathway combined with PD1/PD-L1 inhibitors are summarised.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
41
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
42
|
Tsai YT, Schlom J, Donahue RN. Blood-based biomarkers in patients with non-small cell lung cancer treated with immune checkpoint blockade. J Exp Clin Cancer Res 2024; 43:82. [PMID: 38493133 PMCID: PMC10944611 DOI: 10.1186/s13046-024-02969-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 03/18/2024] Open
Abstract
The paradigm of non-small cell lung cancer (NSCLC) treatment has been profoundly influenced by the development of immune checkpoint inhibitors (ICI), but the range of clinical responses observed among patients poses significant challenges. To date, analyses of tumor biopsies are the only parameter used to guide prognosis to ICI therapy. Tumor biopsies, however, are often difficult to obtain and tissue-based biomarkers are limited by intratumoral heterogeneity and temporal variability. In response, there has been a growing emphasis on the development of "liquid biopsy"‒ derived biomarkers, which offer a minimally invasive means to dynamically monitor the immune status of NSCLC patients either before and/or during the course of treatment. Here we review studies in which multiple blood-based biomarkers encompassing circulating soluble analytes, immune cell subsets, circulating tumor DNA, blood-based tumor mutational burden, and circulating tumor cells have shown promising associations with the clinical response of NSCLC patients to ICI therapy. These investigations have unveiled compelling correlations between the peripheral immune status of patients both before and during ICI therapy and patient outcomes, which include response rates, progression-free survival, and overall survival. There is need for rigorous validation and standardization of these blood-based assays for broader clinical application. Integration of multiple blood-based biomarkers into comprehensive panels or algorithms also has the potential to enhance predictive accuracy. Further research aimed at longitudinal monitoring of circulating biomarkers is also crucial to comprehend immune dynamics and resistance mechanisms and should be used alongside tissue-based methods that interrogate the tumor microenvironment to guide treatment decisions and may inform on the development of novel therapeutic strategies. The data reviewed here reinforce the opportunity to refine patient stratification, optimize treatments, and improve outcomes not only in NSCLC but also in the wider spectrum of solid tumors undergoing immunotherapy.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Le Chapelain O, Jadoui S, Gros A, Barbaria S, Benmeziane K, Ollivier V, Dupont S, Solo Nomenjanahary M, Mavouna S, Rogozarski J, Mawhin MA, Caligiuri G, Delbosc S, Porteu F, Nieswandt B, Mangin PH, Boulaftali Y, Ho-Tin-Noé B. The localization, origin, and impact of platelets in the tumor microenvironment are tumor type-dependent. J Exp Clin Cancer Res 2024; 43:84. [PMID: 38493157 PMCID: PMC10944607 DOI: 10.1186/s13046-024-03001-2] [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: 11/27/2023] [Accepted: 03/01/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND How platelets interact with and influence the tumor microenvironment (TME) remains poorly characterized. METHODS We compared the presence and participation of platelets in the TME of two tumors characterized by highly different TME, PyMT AT-3 mammary tumors and B16F1 melanoma. RESULTS We show that whereas firmly adherent platelets continuously line tumor vessels of both AT-3 and B16F1 tumors, abundant extravascular stromal clusters of platelets from thrombopoietin-independent origin were present only in AT-3 mammary tumors. We further show that platelets influence the angiogenic and inflammatory profiles of AT-3 and B16F1 tumors, though with very different outcomes according to tumor type. Whereas thrombocytopenia increased bleeding in both tumor types, it further caused severe endothelial degeneration associated with massive vascular leakage, tumor swelling, and increased infiltration of cytotoxic cells, only in AT-3 tumors. CONCLUSIONS These results indicate that while platelets are integral components of solid tumors, their localization and origin in the TME, as well as their impact on its shaping, are tumor type-dependent.
Collapse
Affiliation(s)
- Ophélie Le Chapelain
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
| | - Soumaya Jadoui
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | - Angèle Gros
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | - Samir Barbaria
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | | | - Véronique Ollivier
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | - Sébastien Dupont
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
| | - Mialitiana Solo Nomenjanahary
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
| | - Sabrina Mavouna
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
| | - Jasmina Rogozarski
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France
| | - Marie-Anne Mawhin
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | | | - Sandrine Delbosc
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | | | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
| | - Pierre H Mangin
- Université de Strasbourg, Institut National de la Santé et de la Recherche Médicale, Etablissement Français du Sang Grand-Est, Unité Mixte de Recherche-S1255, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg, F-67065, France
| | - Yacine Boulaftali
- Université Paris Cité, INSERM UMR 1148, LVTS, Paris, F-75018, France
| | - Benoit Ho-Tin-Noé
- Faculté de Pharmacie de Paris, Université Paris Cité, Inserm UMR-S 1144 -Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, Paris, 75006, France.
| |
Collapse
|
44
|
Peng C, Wang Y, Zhang H, Chen P. The platelet-related genes associated with the prognosis of HCC by regulating cycling T cell and prolif-TAMs. Heliyon 2024; 10:e26798. [PMID: 38486758 PMCID: PMC10938119 DOI: 10.1016/j.heliyon.2024.e26798] [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: 05/23/2023] [Revised: 01/15/2024] [Accepted: 02/20/2024] [Indexed: 03/17/2024] Open
Abstract
Accumulating evidence highlighted the important roles of platelets in the prognosis and progression of various tumors. Nevertheless, the role of platelet-related genes (PRGs) in HCC remains limited. In this work, 92 differentially expressed PRGs were described in HCC using TCGA and ICGC databases. Then, based on the different expressions of PRGs, we explored two subtypes and developed the PRGs prognostic signature in HCC. The PRGs signature was an independent prognosis factor associated with immune cell infiltration in HCC. Furthermore, two external validation sets verified the expression and prognosis of the PRGs signature gene in HCC. Finally, scRNA-seq analysis demonstrated that the signature genes (CENPE and KIF2C) were mainly expressed in cycling T cells and prolif-TAMs. Enrichment analysis showed that CENPE and KIF2C regulated the cell cycle and p53 pathways in these cells. In conclusion, this study builds the PRGs-related risk signature of HCC and reveals the potential mechanism by which these signature genes regulate the immune microenvironment in HCC.
Collapse
Affiliation(s)
- Chenjia Peng
- School of Mathematics and Computational Science, Hunan First Normal University, Changsha, 410205, PR China
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Ying Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Hengbo Zhang
- Physical Education Department, First Hunan Normal University, Changsha, 410081, PR China
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| |
Collapse
|
45
|
Chen L, Zhang D, Chen Y, Zhu H, Liu Z, Yu Z, Xie J. ORC6 acts as an effective prognostic predictor for non‑small cell lung cancer and is closely associated with tumor progression. Oncol Lett 2024; 27:96. [PMID: 38288041 PMCID: PMC10823314 DOI: 10.3892/ol.2024.14229] [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: 09/26/2023] [Accepted: 12/07/2023] [Indexed: 01/31/2024] Open
Abstract
Origin recognition complexes (ORCs) are vital in the control of DNA replication and the progression of the cell cycle, however the precise function and mechanism of ORC6 in non-small cell lung cancer (NSCLC) is still not well understood. The present study used bioinformatics methods to assess the predictive significance of ORC6 expression in NSCLC. Moreover, the expression of ORC6 was further evaluated using reverse transcription-quantitative PCR and western blotting, and its functional significance in lung cancer was assessed via knockdown experiments using small interfering RNA. A significant association was demonstrated between the expression of ORC6 and the clinical features of NSCLC. In particular, elevated levels of ORC6 were significantly strongly correlated with an unfavorable prognosis. Multivariate analysis demonstrated that increased ORC6 expression independently contributed to the risk of overall survival (HR 1.304; P=0.015) in individuals diagnosed with NSCLC. Analysis of Kaplan-Meier plots demonstrated that ORC6 expression served as a valuable indicator for diagnosing and predicting the prognosis of NSCLC. Moreover, in vitro studies demonstrated that modified ORC6 expression had a significant impact on the proliferation, migration and metastasis of NSCLC cells. NSCLC cell lines (H1299 and mH1650) exhibited markedly higher ORC6 expression than normal lung cell lines. The results of the present study indicated a strong association between the expression of ORC6 and the clinicopathological characteristics of NSCLC, which suggested its potential as a reliable biomarker for predicting NSCLC. Furthermore, ORC6 may have important therapeutic implications in the management of NSCLC.
Collapse
Affiliation(s)
- Letian Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Dongdong Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Yujuan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Huilan Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhipeng Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhiping Yu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Junping Xie
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
46
|
Bi H, Ren D, Xiao Y, Zhou Y, Yi B, Han W, Shao Y, Wang J, Zhang C, Wang H. Prognostic implications of neutrophil-to-lymphocyte ratio in patients with extensive-stage small cell lung cancer receiving chemoimmunotherapy: A multicenter, real-world study. Thorac Cancer 2024; 15:559-569. [PMID: 38294282 PMCID: PMC10912525 DOI: 10.1111/1759-7714.15225] [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: 11/06/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) are closely related to the prognosis of patients with non-small cell lung cancer, but their effect on extensive-stage small cell lung cancer (ES-SCLC) remains uncertain. METHODS This retrospective study was conducted in ES-SCLC patients treated with first-line atezolizumab or durvalumab and platinum-etoposide. Clinical data from three hospitals were analyzed. Significant risk factors for survival were identified using descriptive statistics and Cox regression. Homogeneity was assessed using t-tests or nonparametric tests. Kaplan-Meier analysis revealed an association between high NLR level and median PFS and OS. RESULTS A total of 300 ES-SCLC patients were included in the study. Cox regression analysis revealed that an elevated NLR level after the second treatment cycle (defined as NLRT2) was an independent prognostic factor for survival. Stratifying patients based on median NLRT2 showed significant differences in both PFS (HR: 1.863, 95% CI: 1.62-2.12, p < 0.001) and OS (HR: 2.581, 95% CI: 2.19-3.04, p < 0.001) between NLR ≥ 1.75 and NLR < 1.75 groups. mPFS and mOS were 8.2 versus 6.1 months and 13.7 versus 9.5 months, respectively. NLR was also associated with treatment efficacy and occurrence of irAEs. Further stratification based on NLR and irAEs showed that in the NLR < 1.75 group, patients with irAEs had prolonged mPFS and mOS. In the NLR ≥ 1.75 group, only mPFS showed a significant difference between patients with and without irAEs. CONCLUSION NLRT2 and irAEs can predict the prognosis of ES-SCLC patients with first-line ES-SCLC receiving PD-L1 inhibitors combined with chemotherapy.
Collapse
Affiliation(s)
- Huanhuan Bi
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Department of Respiratory and Critical Care MedicineThe Affiliated Qingdao Central Hospital of Medical College of Qingdao UniversityQingdaoChina
| | - Dunqiang Ren
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yuting Xiao
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yinxue Zhou
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Bingqian Yi
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Weizhong Han
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Yanmei Shao
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Jingluan Wang
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Chunling Zhang
- Department of Respiratory and Critical Care MedicineThe Affiliated Qingdao Central Hospital of Medical College of Qingdao UniversityQingdaoChina
| | - Hongmei Wang
- Department of Respiratory and Critical Care Medicinethe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
47
|
Schöne N, Kemper M, Menck K, Evers G, Krekeler C, Schulze AB, Lenz G, Wardelmann E, Binder C, Bleckmann A. PD-L1 on large extracellular vesicles is a predictive biomarker for therapy response in tissue PD-L1-low and -negative patients with non-small cell lung cancer. J Extracell Vesicles 2024; 13:e12418. [PMID: 38453684 PMCID: PMC10920108 DOI: 10.1002/jev2.12418] [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: 03/31/2023] [Revised: 10/24/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Immunotherapy has revolutionized the treatment of patients with non-small cell lung cancer (NSCLC). High expression of tissue PD-L1 (tPD-L1) is currently the only approved biomarker for predicting treatment response. However, even tPD-L1 low (1-49%) and absent (<1%) patients might benefit from immunotherapy but, to date, there is no reliable biomarker, that can predict response in this particular patient subgroup. This study aimed to test whether tumour-associated extracellular vesicles (EVs) could fill this gap. Using NSCLC cell lines, we identified a panel of tumour-related antigens that were enriched on large EVs (lEVs) compared to smaller EVs. The levels of lEVs carrying these antigens were significantly elevated in plasma of NSCLC patients (n = 108) and discriminated them from controls (n = 77). Among the tested antigens, we focused on programmed cell death ligand 1 (PD-L1), which is a well-known direct target for immunotherapy. In plasma lEVs, PD-L1 was mainly found on a population of CD45- /CD62P+ lEVs and thus seemed to be associated with platelet-derived vesicles. Patients with high baseline levels of PD-L1+ lEVs in blood showed a significantly better response to immunotherapy and prolonged survival. This was particularly true in the subgroup of NSCLC patients with low or absent tPD-L1 expression, thus identifying PD-L1-positive lEVs in plasma as a novel predictive and prognostic marker for immunotherapy.
Collapse
Affiliation(s)
- Nadja Schöne
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Marcel Kemper
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Kerstin Menck
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Georg Evers
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Carolin Krekeler
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Arik Bernard Schulze
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| | - Georg Lenz
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
| | - Eva Wardelmann
- University of Münster, Gerhard‐Domagk‐Institute of PathologyMünsterGermany
| | - Claudia Binder
- University Medicine Göttingen, Clinic for Hematology/Medical OncologyGöttingenGermany
| | - Annalen Bleckmann
- University of Münster, Department of Medicine A, Hematology, Oncology, and PneumologyMünsterGermany
- University Hospital Münster, West German Cancer CenterMünsterGermany
| |
Collapse
|
48
|
Hu Z, Wang N, Zhang Y, Zhang D, Sun S, Yu H, Lin Y, Zhao X, Wang H, Wu X, Ichiki Y, Watanabe S, Gong Z, Chang J, Wang J. PD-L1 mRNA derived from tumor-educated platelets as a potential immunotherapy biomarker in non-small cell lung cancer. Transl Lung Cancer Res 2024; 13:345-354. [PMID: 38496687 PMCID: PMC10938106 DOI: 10.21037/tlcr-24-29] [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: 01/09/2024] [Accepted: 02/15/2024] [Indexed: 03/19/2024]
Abstract
Background To date, the role of programmed death ligand-1 (PD-L1) messenger RNA (mRNA) derived from tumor-educated platelets (TEPs) has not been well investigated in patients with advanced non-small cell lung cancer (NSCLC). A few reports have examined whether mRNA in TEPs can predict the clinical responses of patients with advanced NSCLC following immunotherapy. This study aimed to identify novel biomarkers to improve the clinical benefits and outcomes of NSCLC patients. Methods Advanced NSCLC patients receiving a combination of immunotherapy and chemotherapy, or immunotherapy alone as a first- or second-line treatment at the Fudan University Shanghai Cancer Center were enrolled in this study. All the patients had wild-type epidermal growth factor receptor/anaplastic lymphoma kinase. The patients were enrolled in clinical trials for immune checkpoint inhibitors (ICIs), including nivolumab, pembrolizumab, atezolizumab, durvalumab, tremelimumab, and camrelizumab. Tumoral PD-L1 expression was tested by immunohistochemistry (PD-L1 22C3 pharmDx kit, Agilent, Santa Clara, CA, USA) in archived tissue samples, when available, to calculate the tumor proportion scores (TPSs). RNA and exosomal RNA of blood were isolated before immunotherapy using the Yunying RNA extraction kit (Yunying Medicine, Shanghai, China). The concentration and quality of the RNA was determined using a Qubit fluorometer (Life Technologies, Carlsbad, CA, USA). Finally, we analyzed the predictive value of TEP-derived PD-L1 mRNA expression and association with the level of the tumoral PD-L1 expression. Results In total, 72 patients were enrolled in this study. Most of the patients were male (n=54, 75.0%), had adenocarcinoma (n=49, 68.1%). We found there was no significant correlation between the TEP-derived mRNA of PD-L1 and tumoral PD-L1 expression based on the results of the Pearson Correlation test (r=-0.19, P=0.233). Based on the median of PD-L1 mRNA, 72 patients were divided into a high PD-L1 group and a low PD-L1 group. We found that 19 patients (44.4%) responded to immunotherapy [partial response or progression-free survival (PFS) >6 months] in the high PD-L1 group, but only five patients (13.9%) responded to immunotherapy in the low PD-L1 group (P<0.01). The median PFS of the low PD-L1 group was lower than that of the high PD-L1 group (2.8 vs. 8.3 months, P<0.001). For the patients who were treated with immunotherapy alone (n=64), a similar PFS advantage was observed in the high PD-L1 group (2.8 vs. 8.0 months, P=0.002). Conclusions This article presented the first data on TEP-derived PD-L1 mRNA in advanced NSCLC patients following immunotherapy and showed the potential advantage of using it as the surrogate biomarker for predicting the PFS and overall survival of patients following immunotherapy.
Collapse
Affiliation(s)
- Zhihuang Hu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Na Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yao Zhang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | | | - Si Sun
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Hui Yu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Ying Lin
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Xinmin Zhao
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Huijie Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Xianghua Wu
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| | - Yoshinobu Ichiki
- Department of General Thoracic Surgery, Saitama Medical Center, Hidaka, Japan
| | - Satoshi Watanabe
- Department of Respiratory Medicine and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Jianhua Chang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China
| | - Jialei Wang
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Institute of Thoracic Oncology, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Bandini S, Ulivi P, Rossi T. Extracellular Vesicles, Circulating Tumor Cells, and Immune Checkpoint Inhibitors: Hints and Promises. Cells 2024; 13:337. [PMID: 38391950 PMCID: PMC10887032 DOI: 10.3390/cells13040337] [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: 01/15/2024] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized the treatment of cancer, in particular lung cancer, while the introduction of predictive biomarkers from liquid biopsies has emerged as a promising tool to achieve an effective and personalized therapy response. Important progress has also been made in the molecular characterization of extracellular vesicles (EVs) and circulating tumor cells (CTCs), highlighting their tremendous potential in modulating the tumor microenvironment, acting on immunomodulatory pathways, and setting up the pre-metastatic niche. Surface antigens on EVs and CTCs have proved to be particularly useful in the case of the characterization of potential immune escape mechanisms through the expression of immunosuppressive ligands or the transport of cargos that may mitigate the antitumor immune function. On the other hand, novel approaches, to increase the expression of immunostimulatory molecules or cargo contents that can enhance the immune response, offer premium options in combinatorial clinical strategies for precision immunotherapy. In this review, we discuss recent advances in the identification of immune checkpoints using EVs and CTCs, their potential applications as predictive biomarkers for ICI therapy, and their prospective use as innovative clinical tools, considering that CTCs have already been approved by the Food and Drug Administration (FDA) for clinical use, but providing good reasons to intensify the research on both.
Collapse
Affiliation(s)
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (S.B.); (T.R.)
| | | |
Collapse
|
50
|
Trivanović D, Mojsilović S, Bogosavljević N, Jurišić V, Jauković A. Revealing profile of cancer-educated platelets and their factors to foster immunotherapy development. Transl Oncol 2024; 40:101871. [PMID: 38134841 PMCID: PMC10776659 DOI: 10.1016/j.tranon.2023.101871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Among multiple hemostasis components, platelets hyperactivity plays major roles in cancer progression by providing surface and internal components for intercellular crosstalk as well as by behaving like immune cells. Since platelets participate and regulate immunity in homeostatic and disease states, we assumed that revealing platelets profile might help in conceiving novel anti-cancer immune-based strategies. The goal of this review is to compile and discuss the most recent reports on the nature of cancer-associated platelets and their interference with immunotherapy. An increasing number of studies have emphasized active communication between cancer cells and platelets, with platelets promoting cancer cell survival, growth, and metastasis. The anti-cancer potential of platelet-directed therapy has been intensively investigated, and anti-platelet agents may prevent cancer progression and improve the survival of cancer patients. Platelets can (i) reduce antitumor activity; (ii) support immunoregulatory cells and factors generation; (iii) underpin metastasis and, (iv) interfere with immunotherapy by expressing ligands of immune checkpoint receptors. Mediators produced by tumor cell-induced platelet activation support vein thrombosis, constrain anti-tumor T- and natural killer cell response, while contributing to extravasation of tumor cells, metastatic potential, and neovascularization within the tumor. Recent studies showed that attenuation of immunothrombosis, modulation of platelets and their factors have a good perspective in immunotherapy optimization. Particularly, blockade of intra-tumoral platelet-associated programmed death-ligand 1 might promote anti-tumor T cell-induced cytotoxicity. Collectively, these findings suggest that platelets might represent the source of relevant cancer staging biomarkers, as well as promising targets and carriers in immunotherapeutic approaches for combating cancer.
Collapse
Affiliation(s)
- Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia.
| | - Slavko Mojsilović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia
| | | | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac 34000, Serbia
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia
| |
Collapse
|