1
|
Yue D, Wang R, Zhao Y, Wu B, Li S, Zeng W, Wan S, Liu L, Dai Y, Shi Y, Xu R, Yang Z, Wang X, Zou Y. Investigating the molecular mechanisms between type 1 diabetes and mild cognitive impairment using bioinformatics analysis, with a focus on immune response. Int Immunopharmacol 2024; 142:113256. [PMID: 39340997 DOI: 10.1016/j.intimp.2024.113256] [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: 03/23/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024]
Abstract
The immune system is involved in the development and progression of several diseases. Type 1 diabetes mellitus (T1DM), an autoimmune disorder, influences the progression of several other conditions; however, the link between T1DM and mild cognitive impairment (MCI) remains unclear. This study investigated the underlying immune response mechanisms that contribute to the development and progression of T1DM and MCI. Microarray datasets for MCI (GSE63060) and T1DM (GSE30208) were retrieved from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) were identified using the limma package. To explore the functional implications of these DEGs, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were conducted using ClusterProfiler. Protein-protein interaction networks for the DEGs were constructed using the STRING database and visualized using Cytoscape. The Molecular Complex Detection algorithm was used to analyze DEGs. Immune cell infiltration in patients with T1DM and MCI was analyzed using the xCell method. Gene set enrichment analysis was used to gain in-depth insights into the functional characteristics of T1DM and MCI. Immune-related genes were obtained from the GeneCards and ImmPort databases. Machine learning algorithms were used to identify potential hub genes associated with immunity for T1DM and MCI diagnosis, and the diagnostic value was assessed using the receiver operating characteristic curve. The identified genes were validated using quantitative polymerase chain reaction. In the T1DM and MCI datasets, 610 DEGs showed consistent trends, of which 232 and 378 were upregulated and downregulated, respectively. Immune response analysis revealed significant changes in the immune cells associated with MCI and T1DM. Using immune-related genes, DEGs, and machine learning techniques, we identified CD3D in the MCI and T1DM groups. We observed a gradual decline in the cognitive function of mice with T1DM as the disease progressed. CD3D expression increased with increasing disease severity; CD3D primarily affected CD4+ T cells. This study revealed a complex interaction between T1DM and MCI, providing novel insights into the intricate relationship between immune dysregulation and cognitive impairment and expanding our understanding of these two interconnected disorders. These findings will facilitate the development of therapeutic interventions and identification of potential therapeutic targets.
Collapse
Affiliation(s)
- Dongxu Yue
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Runze Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Yanli Zhao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Bangxu Wu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Shude Li
- Department of Biochemistry and Molecular Biology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Weilin Zeng
- Department of Pathogen Biology and Immunology, Kunming Medical University, Kunming, PR China
| | - Shanshan Wan
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, PR China
| | - Lifang Liu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Yating Dai
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Yuling Shi
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Ruobing Xu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China
| | - Zhihong Yang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China.
| | - Xie Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China.
| | - Yingying Zou
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, PR China.
| |
Collapse
|
2
|
Wang L, He Z, Fan S, Mo L, Li Y, Yuan X, Xu B, Mou Y, Yin Y. Quantitative analysis of immune cells within the tumor microenvironment of glioblastoma and their relevance for prognosis. Int Immunopharmacol 2024; 142:113109. [PMID: 39255678 DOI: 10.1016/j.intimp.2024.113109] [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: 06/20/2024] [Revised: 08/09/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024]
Abstract
Glioblastoma (GBM) is a high malignant tumor with no effective treatment. To comprehensively characterize the landscape of immune cells in GBM and evaluate their correlation with prognosis, we developed a multispectral fluorescent imaging pipeline that included tumor-infiltrating lymphocytic markers (CD3, CD4, CD8, FOXP3, NKP46), immune checkpoint markers (PD-1, PD-L1), and markers to characterize myeloid cells (CD68, CD66b, CD163, HLA-DR), to spatially quantify 18 immune cell subsets in 21 GBM cases. We found that macrophages are the most abundant in GBM microenvironment, followed by T cells and neutrophils, while NK and NKT cells are the least. Previously unreported CD8+ Treg, PD-L1+ neutrophils, and high proportion of PD-1+ NK and PD-1+ T cells were also detected. Single high densities of PD-1+CD8+ T cells, neutrophils, and PD-L1-expressing CD68+ cells were associated with longer survival. Moreover, closer proximity of T cells to PD-L1+ macrophages or PD-L1+ neutrophils were associated with poor prognosis. Correlative analysis revealed circulating PMN-MDSC and e-MDSC were positively correlated with intratumoral M2 macrophages, while circulating NK cells were inversely associated with infiltrating CD4+ Treg cells in GBM patients. Our findings highlighted the potential roles of infiltrating immune cells in prognosis prediction and developing novel immunotherapeutic strategies for GBM patients.
Collapse
Affiliation(s)
- Lu Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Zhenqiang He
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shuning Fan
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Li Mo
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China
| | - Xia Yuan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Yanhui Yin
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-related Diseases, Peking University, Beijing 100191, China.
| |
Collapse
|
3
|
He Y, Wang X. A comprehensive investigation of associations between cell death pathways and molecular and clinical features in pan-cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03769-x. [PMID: 39487950 DOI: 10.1007/s12094-024-03769-x] [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: 09/13/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Regulated cell death (RCD) pathways play significant roles in tumorigenesis. However, systematic investigation into correlations between RCD and various molecular and clinical features, particularly anti-tumor immunity and immunotherapy response in pan-cancer remains lacking. METHODS Using the single-sample gene set enrichment analysis, we quantified the activities of six RCD pathways (apoptosis, autophagy, ferroptosis, cuproptosis, necroptosis, and pyroptosis) in each cancer specimen. Then, we explored associations of these six RCD pathways with tumor immunity, genomic instability, tumor phenotypes and clinical features, and responses to immunotherapy and targeted therapies in pan-cancer by statistical analyses. RESULTS Our results showed that the RCD (except autophagy) activities were oncogenic signatures, as evidenced by their hyperactivation in late stage or metastatic cancer patients, positive correlations with tumor proliferation, stemness, genomic instability and intratumor heterogeneity, and correlation with worse survival outcomes in cancer. In contrast, autophagy was a tumor suppressive signature as its associations with molecular and clinical features in cancer shows an opposite pattern compared to the other RCD pathways. Furthermore, heightened RCD (except cuproptosis) activities were correlated with increased sensitivity to immune checkpoint inhibitors. Additionally, elevated activities of pyroptosis, autophagy, cuproptosis and necroptosis were associated with increased drug sensitivity in a broad spectrum of anti-tumor targeted therapies, while the elevated activity of ferroptosis was correlated with decreased sensitivity to numerous targeted therapies. CONCLUSION RCD (except autophagy) activities correlate with unfavorable cancer prognosis, while the autophagy activity correlate with favorable clinical outcomes. RCD (except cuproptosis) activities are positive biomarkers for anti-tumor immunity and immunotherapy response.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
- Intelligent Pharmacy Interdisciplinary Research Center, China Pharmaceutical University, Nanjing, 211198, China.
- Big Data Research Institute, China Pharmaceutical University, Nanjing, 211198, China.
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
4
|
Kizil B, De Virgiliis F, Scheiermann C. Neural control of tumor immunity. FEBS J 2024; 291:4670-4679. [PMID: 39304984 DOI: 10.1111/febs.17280] [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: 02/23/2024] [Revised: 06/02/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024]
Abstract
Communication between the nervous system and the immune system has evolved to optimally respond to potentially dangerous stimuli both from within and outside the body. Tumors pose a severe threat to an organism and current therapies are insufficient for tumor regression in the majority of cases. Studies show that tumors are innervated by peripheral nerves from the sensory, parasympathetic and sympathetic nervous systems. Interactions between cancer cells, nerves and immune cells regulate overall tumor progression. Clinical studies have indicated the potential of targeting the peripheral nervous system for promoting anti-tumor immune responses. This view point provides an opinion on the current evidence and therapeutic potential of manipulating neuro-immune communications in cancer.
Collapse
Affiliation(s)
- Burak Kizil
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Francesco De Virgiliis
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Christoph Scheiermann
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
- Geneva Center for Inflammation Research (GCIR), Geneva, Switzerland
- Translational Research Centre in Onco-Hematology (CRTOH), Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Switzerland
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel-Center for Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Planegg-Martinsried, Germany
| |
Collapse
|
5
|
Sancho-Araiz A, Parra-Guillen ZP, Troconiz IF, Freshwater T. Disentangling Anti-Tumor Response of Immunotherapy Combinations: A Physiologically Based Framework for V937 Oncolytic Virus and Pembrolizumab. Clin Pharmacol Ther 2024; 116:1304-1313. [PMID: 39037559 DOI: 10.1002/cpt.3379] [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: 03/03/2024] [Accepted: 07/04/2024] [Indexed: 07/23/2024]
Abstract
Immuno-oncology (IO) is a growing strategy in cancer treatment. Oncolytic viruses (OVs) can selectively infect cancer cells and lead to direct and/or immune-dependent tumor lysis. This approach represents an opportunity to potentiate the efficacy of immune checkpoint inhibitors (ICI), such as pembrolizumab. Currently, there is a lack of comprehensive quantitative models for the aforementioned scenarios. In this work, we developed a mechanistic framework describing viral kinetics, viral dynamics, and tumor response after intratumoral (i.t.) or intravenous (i.v.) administration of V937 alone or in combination with pembrolizumab. The model accounts for tumor shrinkage, in both injected and non-injected lesions, induced by: viral-infected tumor cell death and activated CD8 cells. OV-infected tumor cells enhanced the expansion of CD8 cells, whereas pembrolizumab inhibits their exhaustion by competing with PD-L1 in their binding to PD-1. Circulating viral levels and treatment effects on tumor volume were adequately characterized in all the different scenarios. This mechanistic-based model has been developed by combining top-down and bottom-up approaches and provides individual estimates of viral and ICI responses. The robustness of the model is reflected by the description of the tumor size time profiles in a variety of clinical scenarios. Additionally, this platform allows us to investigate not only the contribution of processes related to the viral kinetics and dynamics on tumor response, but also the influence of its interaction with an ICI. Additionally, the model can be used to explore different scenarios aiming to optimize treatment combinations and support clinical development.
Collapse
Affiliation(s)
- Aymara Sancho-Araiz
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Zinnia P Parra-Guillen
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Iñaki F Troconiz
- Department of Pharmaceutical Science, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Institute of Data Science and Artificial Intelligence, DATAI, University of Navarra, Pamplona, Spain
| | - Tomoko Freshwater
- Oncology Early Development, Clinical Research, Merck & Co., Inc., Rahway, New Jersey, USA
| |
Collapse
|
6
|
Yan T, Zhou W, Li C. Discovery of a T cell proliferation-associated regulator signature correlates with prognosis risk and immunotherapy response in bladder cancer. Int Urol Nephrol 2024; 56:3447-3462. [PMID: 38789872 DOI: 10.1007/s11255-024-04086-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND The efficacy of immunotherapy is heavily influenced by T cell activity. This study aimed to examine how T cell proliferation regulators can predict the prognosis and response to immunotherapy in patients with bladder cancer (BCa). METHODS T cell proliferation-related subtypes were determined by employing the non-negative matrix factorization (NMF) algorithm that analyzed the expression patterns of T cell proliferation regulators. Subtypes were assessed for variations in prognosis, immune infiltration, and functional behaviors. Subsequently, a risk model related to T cell proliferation was created through Cox and Lasso regression analyses in the TCGA cohort and then confirmed in two GEO cohorts and an immunotherapy cohort. RESULTS BCa patients were categorized into two subtypes (C1 and C2) according to the expression profiles of 31 T cell proliferation-related genes (TRGs) with distinct prognoses and immune landscapes. The C2 subtype had a shorter overall survival (OS), with higher levels of M2 macrophage infiltration, and the activation of cancer-related pathways than the C1 subtype. Following this, thirteen prognosis-related genes that were involved in T cell proliferation were utilized to create the prognostic signature. The model's predictive accuracy was confirmed by analyzing both internal and external datasets. Individuals in the high-risk category experienced a poorer prognosis, increased immunosuppressive factors in the tumor microenvironment, and diminished responses to immunotherapy. Additionally, the immunotherapeutic prediction efficacy of the model was further confirmed by an immunotherapy cohort (anti-PD-L1 in the IMvigor210 cohort). CONCLUSIONS Our study characterized two subtypes linked to T cell proliferation in BCa patients with distinct prognoses and tumor microenvironment (TME) patterns, providing new insights into the heterogeneity of T cell proliferation in BCa and its connection to the immune landscape. The signature has prospective clinical implications for predicting outcomes and may help physicians to select prospective responders who prioritize current immunotherapy.
Collapse
Affiliation(s)
- Ting Yan
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China
| | - Wei Zhou
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, People's Republic of China
| | - Chun Li
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China.
| |
Collapse
|
7
|
Walter N, Bärtl S, Alt V, Rupp M. Recent advancements and future directions in fracture related infections: A scoping review. Injury 2024; 55 Suppl 6:111902. [PMID: 39482033 DOI: 10.1016/j.injury.2024.111902] [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: 03/21/2024] [Revised: 08/29/2024] [Accepted: 09/15/2024] [Indexed: 11/03/2024]
Abstract
Fracture-related infections (FRIs) are complex challenges in orthopedic and trauma surgery, driving ongoing advancements in diagnostics, therapeutics, and management strategies. This scoping review examines recent progress and future directions in FRI management. Diagnostic enhancements encompass standardized definitions, improved biomarkers, advanced microbiological techniques, and innovative imaging modalities. Promising future diagnostics may include point-of-care testing, advanced imaging with enhanced specificity, and machine learning algorithms. Advancements in implant technology emphasize materials science, surface modifications, and personalized 3D printing, enhancing durability and antimicrobial efficacy. Immunomodulatory therapies targeting T cell dysfunction offer potential in addressing FRI chronicity. Enzybiotics and phages present promising alternatives to combat antibiotic resistance, with enzybiotics demonstrating effectiveness against biofilm-associated infections. Patient optimization, multidisciplinary approaches and specialized reference centers play vital roles in comprehensive FRI management, particularly crucial in resource-constrained settings. Collaboration and investment in research and technology are imperative for harnessing the full potential of these advancements and improving global FRI management outcomes. Addressing these complexities necessitates a multifaceted approach integrating clinical expertise, technological innovation, and global cooperation to optimize patient care and mitigate the burden of FRI worldwide.
Collapse
Affiliation(s)
- Nike Walter
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany.
| | - Susanne Bärtl
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Volker Alt
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Markus Rupp
- Department for Trauma Surgery, University Hospital Regensburg, Franz- Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| |
Collapse
|
8
|
Xiong J, Ouyang W, Yang M, Gao Z, Zhou H, Lou H, Guo Y, Xu Z, Zheng L, Liu Y, Wang Z, Sun P, Niyazi H, Wang J, Chen Y, Zhang B, Li L, Kang X, Guo W. Efficacy and Safety of Iparomlimab, an Anti-PD-1 Antibody, in Patients with Advanced Solid Tumors: A Phase 1c Study. Adv Ther 2024; 41:4153-4171. [PMID: 39276185 DOI: 10.1007/s12325-024-02981-z] [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: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024]
Abstract
INTRODUCTION Iparomlimab (QL1604) is a humanized immunoglobulin G4 mAb against programmed cell death protein 1 (PD-1). Here, we report the preliminary efficacy, safety, pharmacokinetics, and immunogenicity of iparomlimab in patients with advanced solid tumors. METHODS In this open-label, phase 1c study, patients with advanced or metastatic solid tumors, either failed or had no standard therapies available, were enrolled and received intravenous iparomlimab at 3 mg/kg once every 3 weeks. The primary efficacy endpoint was the objective response rate (ORR) assessed by the investigator per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. RESULTS Between July 20, 2020, and September 6, 2021, 71 patients were enrolled and received at least one dose of iparomlimab. The ORR was 9.9% (7/71) and disease control rate was 36.6% (26/71). Median duration of response of all responders was 10.7 months [95% confidence interval (CI), 1.4-not estimable]. Additionally, the median time to progression, progression-free survival, and overall survival were 1.4 months (95% CI, 1.4-2.8), 1.4 months (95% CI, 1.4-2.7), and 9.7 months (95% CI, 7.2-15.3), respectively. A total of 52 (73.2%) patients experienced treatment-related adverse events (TRAEs) (grade ≥ 3, 19.7%). The most common TRAE (≥ 10%) was anemia (18.3%). A total of 20 (28.2%) experienced immune-related adverse events (grade ≥ 3, 7.0%). TRAEs leading to discontinuation of study drug occurred in 4 (5.6%) patients, including immune-mediated myocarditis (2 patients), Guillain-Barré syndrome (1 patient), and diarrhea (1 patient). CONCLUSIONS Iparomlimab showed preliminary clinical activity and had a manageable safety profile in patients with advanced solid tumors. These results support further investigation of iparomlimab as monotherapy or in combination therapy in advanced solid tumors. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT05801094. Retrospectively registered in 2023-03-24.
Collapse
Affiliation(s)
- Jianping Xiong
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiwei Ouyang
- Phase I Ward, Guizhou Cancer Hospital, Guiyang, China
| | - Mengxiang Yang
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, China
| | - Zhenyuan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huan Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hanmei Lou
- Phase I Ward, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yabing Guo
- Liver Cancer Center/Phase I Clinical Research Laboratory, Nanfang Hospital, Guangzhou, China
| | - Zhongyuan Xu
- Liver Cancer Center/Phase I Clinical Research Laboratory, Nanfang Hospital, Guangzhou, China
| | - Ling Zheng
- Phase I Ward, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Ying Liu
- The Third Ward of Digestive Diseases, Henan Cancer Hospital, Zhengzhou, China
| | - Zhongfeng Wang
- Henan Cancer Hospital, The First Hospital of Jilin University, Changchun, China
| | - Ping Sun
- Department of Medical Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Huerxidan Niyazi
- Department of Oncology/Phase I Ward, The First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, China
| | - Jianhua Wang
- Department of Oncology/Phase I Ward, The First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, China
| | - Yan Chen
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Baihui Zhang
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Lingyan Li
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Xiaoyan Kang
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
9
|
Gu Y, Zhao Q. Clinical Progresses and Challenges of Bispecific Antibodies for the Treatment of Solid Tumors. Mol Diagn Ther 2024; 28:669-702. [PMID: 39172329 PMCID: PMC11512917 DOI: 10.1007/s40291-024-00734-w] [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] [Accepted: 07/26/2024] [Indexed: 08/23/2024]
Abstract
In recent years, bispecific antibodies (BsAbs) have emerged as a promising therapeutic strategy against tumors. BsAbs can recruit and activate immune cells, block multiple signaling pathways, and deliver therapeutic payloads directly to tumor sites. This review provides a comprehensive overview of the recent advances in the development and clinical application of BsAbs for the treatment of solid tumors. We discuss the different formats, the unique mechanisms of action, and the clinical outcomes of the most advanced BsAbs in solid tumor therapy. Several studies have also analyzed the clinical progress of bispecific antibodies. However, this review distinguishes itself by exploring the challenges associated with bispecific antibodies and proposing potential solutions. As the field progresses, BsAbs hold promise to redefine cancer treatment paradigms and offer new hope to patients with solid tumors.
Collapse
Affiliation(s)
- Yuheng Gu
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China.
| |
Collapse
|
10
|
Watts T, Jennings S, Anstey S, Roche D. Supporting People Treated with Immune Checkpoint Inhibitors: A Qualitative Study Exploring Oncology Healthcare Professionals' Experiences. Semin Oncol Nurs 2024:151745. [PMID: 39482207 DOI: 10.1016/j.soncn.2024.151745] [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: 08/30/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVES Immune checkpoint inhibitors have recently developed successfully in treatment for several advanced cancers, including advanced renal cancer, where options have previously been limited. However, while some are able to tolerate these treatments, others may experience unpredictable and sometimes severe immune-related adverse events. Oncology health care professionals have vital roles in optimizing safety and supporting positive outcomes for people receiving these treatments. This study aimed to better understand these professionals' experiences of supporting people receiving immune checkpoint inhibitors. METHODS A qualitative exploratory methodology was adopted using semi-structured interviews with 18 purposively sampled senior oncology health professionals, including 12 nurses, who had experience caring for people being treated with checkpoint inhibitors. Data were collected between June and September 2020, transcribed verbatim, and analyzed using reflexive thematic analysis. RESULTS The analysis identified three main themes: First, participants were positive about the potential benefits that checkpoint inhibitors afforded patients, balanced against challenges associated with ambiguities of the treatments and potential impact on existing workloads. Secondly, participants identified the importance of proactive patient monitoring for early detection and reporting of adverse events. Participants highlighted potential challenges if these events went undetected, particularly in the context of the expectation for patient recognition and prompt reporting. Finally, participants identified the need for continual enhancement of health professionals' knowledge and understanding of immunotherapy, supported by the prioritizing of formal immunotherapy education. CONCLUSIONS Whilst immune checkpoint inhibitors offer the possibility for improved disease outcomes, this is balanced against uncertainties regarding potentially unpredictable, often complex, adverse treatment events. This study shows that nurses have vital roles in supporting people receiving these treatments. IMPLICATIONS FOR NURSING PRACTICE Effective care and treatment management for people receiving checkpoint inhibitors require nurses' support through their expert knowledge of immunotherapy and their skills for appropriate coordination and organization of cross-boundary care.
Collapse
Affiliation(s)
- Tessa Watts
- Reader, School of Healthcare Sciences, Cardiff University, Cardiff, Wales.
| | - Stephen Jennings
- Lecturer, Bristol Medical School, Bristol University, Cardiff, Wales
| | - Sally Anstey
- Reader Emirata, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| | - Dominic Roche
- Senior Lecturer, School of Healthcare Sciences, Cardiff University, Cardiff, Wales
| |
Collapse
|
11
|
Guo Y, Shen B, Lou C, Wang L, Li Y. IGSF1: a biomarker for predicting prognosis, immunotherapy response, and drug candidates in COVID-19 combined hepatocellular carcinoma. Discov Oncol 2024; 15:599. [PMID: 39470901 PMCID: PMC11522225 DOI: 10.1007/s12672-024-01483-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly heterogeneous malignancy with poor prognosis and a common cause of cancer-related death worldwide, and despite ongoing therapeutic breakthroughs, patient survival benefits are limited. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of coronavirus disease 2019 (COVID-19) and poses a major threat to humanity worldwide. As the epidemic continues to develop, more and more people are infected with SARS-CoV-2, including patients with HCC. However, the relationship between COVID-19 and HCC has not yet been fully elucidated. Our study aimed to identify the shared genetic characteristics and molecular mechanisms between COVID-19 and HCC. The data involved in this study come from Gene Expression Omnibus (GEO), The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression(GTEx), and Cancer Cell Line Encyclopedia(CCLE) databases. We used differentially expressed genes to perform enrichment analysis to reveal the biological landscape of COVID-19 combined with HCC. In addition, weighted gene co-expression network analysis (WGCNA) was used to study the co-expression network related to COVID-19 and HCC. We then combined the validation datasets to screen out immunoglobulin superfamily member 1 (IGSF1) as the most important core gene. Finally, we extensively studied the functional expression of IGSF1 in tumor samples, normal tissues, and cancer cell lines. The molecular mechanisms related to COVID-19 and HCC are rarely studied. Our study identifies IGSF1 as a potential therapeutic target and immune-related biomarker for patients with COVID-19 and HCC.
Collapse
Affiliation(s)
- Yuanhui Guo
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Baixuan Shen
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Chaoxuan Lou
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Li Wang
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Ying Li
- Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
12
|
Liu D, Tong H, Guo Y, Liu B, Ye C, Yang N, Wu Y. The Toll-like receptor 4 antagonist TAK-242 in combination with sodium hyaluronate alleviates postoperative abdominal adhesion in a mouse model. BMC Med Genomics 2024; 17:257. [PMID: 39456047 PMCID: PMC11520138 DOI: 10.1186/s12920-024-02031-1] [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: 08/15/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Postoperative abdominal adhesion is one of the most common complications after abdominal surgery. The Toll-like receptor 4 (TLR4) signaling pathway is one of the most common inflammation-related pathways, and it has been demonstrated that TLR4 is highly expressed in adhesive tissues; however, the function of TLR4 in adhesion formation has not yet been studied. In the present study, the expression of TLR4 was first detected by immunohistochemical (IHC) and double-immunofluorescence staining in 40 mice, which were randomly divided into four groups, and sacrificed at 1, 3, 5 and 7 days after surgery. Subsequently, another 40 mice were randomly divided into five groups; with the exception of the sham group, the other groups were modeled and treated with saline that contained DMSO, sodium hyaluronate (HA), TAK-242 or TAK-242 + HA (applied to damaged peritoneal wounds). A total of 7 days after surgery, the mice were sacrificed and specimens were collected. Inflammation was detected by hematoxylin and eosin staining, and ELISA of transforming growth factor- β1 (TGF-β1) and interleukin-6 (IL-6); collagen deposition was examined by Masson staining and IHC staining of α-SMA; and reactive oxygen species (ROS) were detected by ROS staining and malondialdehyde (MDA) assay. The results revealed that TLR4 was highly expressed in the adhesive tissues at 3, 5 and 7 days after surgery. In addition, TAK-242 + HA treatment could reduce abdominal adhesion formation, exhibiting lower Nair's score and inflammation scores, lower TGF-β1 and IL-6 levels, and lower collagen thickness and α-SMA levels compared with those in the control group. In addition, the TAK-242 + HA group had lower levels of ROS and MDA compared with those in the control group. The present study revealed that TLR4 was highly expressed in the process of adhesion formation and its inhibitor, TAK-242, combined with HA, could reduce adhesion formation by reducing inflammation and ROS, and alleviating collagen deposition.
Collapse
Affiliation(s)
- Dong Liu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Haochongyang Tong
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Yu Guo
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Bin Liu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Changchun Ye
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, P.R. China
| | - Ni Yang
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Yunhua Wu
- The Second Department of General Surgery, Shaanxi Provincial People's Hospital, 256 West Youyi Road, Xi'an, 710061, Shaanxi, P.R. China.
| |
Collapse
|
13
|
Zhou L, Zhang W, Hu X, Wang D, Tang D. Metabolic Reprogramming of Cancer-Associated Fibroblast in the Tumor Microenvironment: From Basics to Clinic. Clin Med Insights Oncol 2024; 18:11795549241287058. [PMID: 39450056 PMCID: PMC11500237 DOI: 10.1177/11795549241287058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/09/2024] [Indexed: 10/26/2024] Open
Abstract
Metabolic reprogramming occurs when tumor cells replenish themselves with nutrients required for growth to meet their metabolic needs. Cancer-associated fibroblasts (CAFs) are activated fibroblasts involved in building the c (TME) to promote tumor progression and metastasis. Metabolic reprogramming of CAFs can interact with cancer cells to generate metabolic crosstalk. Furthermore, CAF metabolic reprogramming has great potential as a new field of tumor treatment. This review summarizes the role of CAFs in TME and the mechanisms by which metabolic reprogramming of CAFs causes cancer progression and metastasis, demonstrating the great potential of CAF metabolic reprogramming in cancer chemotherapy and immunotherapy treatment. Furthermore, we provide an outlook for future CAF metabolic reprogramming for cancer treatment.
Collapse
Affiliation(s)
- Lujia Zhou
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenjie Zhang
- Department of Clinical Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoxue Hu
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Department of General Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Department of General Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, China
| |
Collapse
|
14
|
Yunyun Z, Guihu W, An J. Explore the expression of mitochondria-related genes to construct prognostic risk model for ovarian cancer and validate it, so as to provide optimized treatment for ovarian cancer. Front Immunol 2024; 15:1458264. [PMID: 39478854 PMCID: PMC11521951 DOI: 10.3389/fimmu.2024.1458264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/17/2024] [Indexed: 11/02/2024] Open
Abstract
Background The use of gene development data from public database has become a new starting point to explore mitochondrial related gene expression and construct a prognostic prediction model of ovarian cancer. Methods Data were obtained from the TCGA and ICGC databases, and the intersection with mitochondrial genes was used to obtain the differentially expressed genes. q-PCR, Cox proportional risk regression, minimal absolute contraction and selection operator regression analysis were performed to construct the prognostic risk model, and ROC curve was used to evaluate the model for centralized verification. The association between risk scores and clinical features, tumor mutation load, immune cell infiltration, macrophage activation analysis, immunotherapy, and chemosensitivity was further evaluated. Results A prognostic risk score model for ovarian cancer patients was constructed based on 12 differentially expressed genes. The score was highly correlated with ovarian cancer macrophage infiltration and was a good predictor of the response to immunotherapy. M1 and M2 macrophages in the ovarian tissue in the OV group were significantly activated, providing a reference for the study of the polarity change of tumor-related macrophages for the prognosis and treatment of ovarian cancer. In terms of drug sensitivity, the high-risk group was more sensitive to vinblastine, Acetalax, VX-11e, and PD-0325901, while the low-risk group was more sensitive to Sabutoclax, SB-505124, cisplatin, and erlotinib. Conclusion The prognostic risk model of ovarian cancer associated to mitochondrial genes built on the basis of public database better evaluated the prognosis of ovarian cancer patients and guided individual treatment.
Collapse
Affiliation(s)
- Zheng Yunyun
- Department of Hepatobiliary Pancreas Surgery and Liver Transplantation, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of AFM (Air Force Medical University), Xi’an, Shaanxi, China
| | - Wang Guihu
- Department of Hepatobiliary Pancreas Surgery and Liver Transplantation, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Jiang An
- Department of Hepatobiliary Pancreas Surgery and Liver Transplantation, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
- National and Local Joint Engineering Research Center of Biodiagnostics and Biotherapy, Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
15
|
Milutinovic S, Jancic P, Jokic V, Petrovic M, Dumic I, Rodriguez AM, Tanasijevic N, Begosh-Mayne D, Stanojevic D, Escarcega RO, Lopez-Mattei J, Cao X. Pembrolizumab-Associated Cardiotoxicity: A Retrospective Analysis of the FDA Adverse Events Reporting System. Pharmaceuticals (Basel) 2024; 17:1372. [PMID: 39459012 PMCID: PMC11510316 DOI: 10.3390/ph17101372] [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: 08/02/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been successfully used in the previous decade for the treatment of a variety of malignancies. Adverse events (AEs) can cause many symptoms, most notably cardiac. We analyzed the frequency of these adverse events, comparing pembrolizumab and other ICIs. METHODS Using the Food and Drug Administration (FDA) adverse event reporting database (FAERS), we searched for all adverse events of interest reported for every ICI included in this study. After obtaining the data, we conducted a disproportionality analysis using the reporting odds ratio (ROR) and the information component (IC). RESULTS A total of 6719 ICI-related cardiac adverse events of interest were reported in the database. Serious outcomes were reported in 100% of the cases, with 34.3% of the cases ending fatally. Compared with all other medications in the database, pembrolizumab use was more frequently associated with myocarditis, pericardial disease, heart failure, and atrial fibrillation. No difference was found in cardiotoxicity between different ICIs. CONCLUSIONS Although infrequent, cardiac AEs in pembrolizumab use are associated with serious outcomes and high mortality. Prospective studies are needed to further research the connection between ICI use and cardiotoxicity.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | - Predrag Jancic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vera Jokic
- Montefiore New Rochelle Hospital, New Rochelle, NY 10801, USA
| | - Marija Petrovic
- Cardiology Fellowship Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Igor Dumic
- Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54703, USA
| | - Ambar Morales Rodriguez
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | | | - Dustin Begosh-Mayne
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | - Dragana Stanojevic
- Clinic for Cardiology, University Clinical Center Nis, 18000 Nis, Serbia
| | - Ricardo O. Escarcega
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
- Lee Health Heart Institute, Fort Myers, FL 33908, USA
| | | | - Xiangkun Cao
- Lee Health Heart Institute, Fort Myers, FL 33908, USA
| |
Collapse
|
16
|
Wang Y, Zhao Y, Zhang G, Lin Y, Fan C, Wei H, Chen S, Guan L, Liu K, Yu S, Fu L, Zhang J, Yuan Y, He J, Cai H. Pan-cancer and single-cell analysis reveal dual roles of lymphocyte activation gene-3 (LAG3) in cancer immunity and prognosis. Sci Rep 2024; 14:24203. [PMID: 39406840 PMCID: PMC11480387 DOI: 10.1038/s41598-024-74808-4] [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: 09/01/2023] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Lymphocyte activating gene-3 (LAG3) is a distinctive T cell co-receptor that is expressed on the surface of lymphocytes. It plays a special inhibitory immune checkpoint role due to its unique domain and signaling pattern. Our aim is to explore the correlation between LAG3 in cancers and physiological processes related to a range of cancers, as well as build LAG3-related immunity and prognostic models. By comprehensively using of datasets and methods from TCGA, GTE-x and GEO databases, cBioPortal, HPA, Kaplan-Meier Plotter, Spearman, CellMinerTM, we delved deeper into the potential impact of the LAG3 in cancer development. These include expression differences, Localization of tumor cell subsets, immune infiltration, matrix infiltration, gene mutations, DNA methylation, signaling pathways and prognosis. Furthermore, we explored LAG3 interactions with different drugs. LAG3 is highly expressed in ACC (p < 0.001), BRCA (p < 0.001), DLBC (p < 0.001), ESCA (p < 0.001), GBM (p < 0.001), HNSC (p < 0.001), KIRC (p < 0.001), LGG (p < 0.001), LUAD (p < 0.01), LUSC (p < 0.001), PAAD (p < 0.001), PCPG (p < 0.01), SKCM (p < 0.001), STAD (p < 0.001), TGCT (p < 0.001) and THCA (p < 0.05), while lowly expressed in COAD (p < 0.001), LIHC (p < 0.05), OV (p < 0.001), PRAD (p < 0.001), READ (p < 0.001), UCEC (p < 0.001) and UCS (p < 0.001). High expression of LAG3 correlates with longer overall survival (OS) in BLCA (HR = 0.67, p < 0.05), CESC (HR = 0.3, p < 0.001), HNSC (HR = 0.67, p < 0.01), LUSC (HR = 0.71, p < 0.05), OV (HR = 0.65, p < 0.01), STAD (HR = 0.68, p < 0.05), and UCEC (HR = 0.57, p < 0.01). Conversely, in KIRC (HR = 1.85, p < 0.001), KIRP (HR = 2.81, p < 0.001), and THYM (HR = 8.92, p < 0.001), high LAG3 expression corresponds to shorter OS. Comprehensive results for recurrence-free survival (RFS) indicate that LAG3 acts as a protective factor in BLCA, CESC, OV, and UCEC. Moreover, LAG3 is widely expressed in tumor-associated lymphocytes, positively correlating with tumor immune scores and stromal scores, and significantly present in the C2 immune subtype across various tumors. High LAG3 expression correlates with increased immune infiltration. LAG3 shows associations with MSI, TMB, and the MMR system, participating in multiple signaling pathways including the T cell receptor pathway. It also demonstrates positive correlations with sensitivity to eleven different drugs. Unlike traditional inhibitory immune checkpoints, LAG3 exhibits dual roles in clinical and immune prognostication across pan-cancers, making it a significant predictive factor. In some cancers, LAG3 serves as a risk factor, indicating adverse clinical outcomes. Conversely, in BLCA, CESC, OV, and UCEC, LAG3 acts as a protective factor associated with longer patient survival. LAG3 demonstrates strong associations within tumor immunity, participating in a range of immune and inflammatory signaling pathways. Elevated levels of LAG3 are linked not only to T cell exhaustion but also to increased immune infiltration and polarization towards M1 macrophages.
Collapse
Affiliation(s)
- Yongfeng Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, 730000, Gansu, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, China
| | - Yanzong Zhao
- School of Stomatology, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Guangming Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yifeng Lin
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Chunling Fan
- School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Hui Wei
- School of Stomatology, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Shude Chen
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Ling Guan
- School of Stomatology, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Kan Liu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Shenhan Yu
- School of Stomatology, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Liangyin Fu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, 730000, Gansu, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, China
| | - Jing Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yuan Yuan
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, 730000, Gansu, China.
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, China.
| | - Jin He
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China.
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, 730000, Gansu, China.
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, China.
| | - Hui Cai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, China.
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, Gansu, China.
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, 204 Donggang West Road, Lanzhou, 730000, Gansu, China.
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu, China.
| |
Collapse
|
17
|
Xiao N, Liu H, Zhang C, Chen H, Li Y, Yang Y, Liu H, Wan J. Applications of single-cell analysis in immunotherapy for lung cancer: Current progress, new challenges and expectations. J Adv Res 2024:S2090-1232(24)00462-4. [PMID: 39401694 DOI: 10.1016/j.jare.2024.10.008] [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: 02/04/2024] [Revised: 06/28/2024] [Accepted: 10/11/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Lung cancer is a prevalent form of cancer worldwide, presenting a substantial risk to human well-being. Lung cancer is classified into two main types: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). The advancement of tumor immunotherapy, specifically immune checkpoint inhibitors and adaptive T-cell therapy, has encountered substantial obstacles due to the rapid progression of SCLC and the metastasis, recurrence, and drug resistance of NSCLC. These challenges are believed to stem from the tumor heterogeneity of lung cancer within the tumor microenvironment. AIM OF REVIEW This review aims to comprehensively explore recent strides in single-cell analysis, a robust sequencing technology, concerning its application in the realm of tumor immunotherapy for lung cancer. It has been effectively integrated with transcriptomics, epigenomics, genomics, and proteomics for various applications. Specifically, these techniques have proven valuable in mapping the transcriptional activity of tumor-infiltrating lymphocytes in patients with NSCLC, identifying circulating tumor cells, and elucidating the heterogeneity of the tumor microenvironment. KEY SCIENTIFIC CONCEPTS OF REVIEW The review emphasizes the paramount significance of single-cell analysis in mapping the immune cells within NSCLC patients, unveiling circulating tumor cells, and elucidating the tumor microenvironment heterogeneity. Notably, these advancements highlight the potential of single-cell analysis to revolutionize lung cancer immunotherapy by characterizing immune cell fates, improving therapeutic strategies, and identifying promising targets or prognostic biomarkers. It is potential to unravel the complexities within the tumor microenvironment and enhance treatment strategies marks a significant step towards more effective therapies and improved patient outcomes.
Collapse
Affiliation(s)
- Nan Xiao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongyang Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Chenxing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Huanxiang Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yang Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ying Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Hongchun Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
18
|
He Y, Ren T, Ji C, Zhao L, Wang X. The baseline hemoglobin level is a positive biomarker for immunotherapy response and can improve the predictability of tumor mutation burden for immunotherapy response in cancer. Front Pharmacol 2024; 15:1456833. [PMID: 39415833 PMCID: PMC11480016 DOI: 10.3389/fphar.2024.1456833] [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: 06/29/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Purpose Because only a subset of cancer patients can benefit from immunotherapy, identifying predictive biomarkers of ICI therapy response is of utmost importance. Methods We analyzed the association between hemoglobin (HGB) levels and clinical outcomes in 1,479 ICIs-treated patients across 16 cancer types. We explored the dose-dependent associations between HGB levels and survival and immunotherapy response using the spline-based cox regression analysis. Furthermore, we investigated the associations across subgroups of patients with different clinicopathological characteristics, treatment programs and cancer types using the bootstrap resampling method. Results HGB levels correlated positively with clinical outcomes in cancer patients receiving immunotherapy but not in those without immunotherapy. Moreover, this association was independent of other clinicopathological characteristics (such as sex, age, tumor stage and tumor mutation burden (TMB)), treatment program and cancer type. Also, this association was independent of the established biomarkers of immunotherapy response, including TMB, PD-L1 expression and microsatellite instability. The combination of TMB and HGB level are more powerful in predicting immunotherapy response than TMB alone. Multi-omics analysis showed that HGB levels correlated positively with antitumor immune signatures and negatively with tumor properties directing antitumor immunosuppression, such as homologous recombination defect, stemness and intratumor heterogeneity. Conclusion The HGB measure has the potential clinical value as a novel biomarker of immunotherapy response that is easily accessible from clinically routine examination. The combination of TMB and HGB measures have better predictive performance for immunotherapy response than TMB.
Collapse
Affiliation(s)
- Yin He
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| | - Tong Ren
- Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Chengfei Ji
- Beijing Highthink Pharmaceutical Technology Service Co., Ltd., Beijing, China
| | - Li Zhao
- Public Experimental Platform, China Pharmaceutical University, Nanjing, China
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Big Data Research Institute, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
19
|
Kim HY, Cho S, Kim SB, Song EC, Jung W, Shin YG, Suh JH, Choi J, Yoon I, Kim U, Ban H, Hwang S, Mun J, Park J, Kim N, Lee Y, Kim MH, Kim S. Specific targeting of cancer vaccines to antigen-presenting cells via an endogenous TLR2/6 ligand derived from cysteinyl-tRNA synthetase 1. Mol Ther 2024; 32:3597-3617. [PMID: 39066478 PMCID: PMC11489552 DOI: 10.1016/j.ymthe.2024.07.014] [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: 01/05/2024] [Revised: 05/16/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer vaccines have been developed as a promising way to boost cancer immunity. However, their clinical potency is often limited due to the imprecise delivery of tumor antigens. To overcome this problem, we conjugated an endogenous Toll-like receptor (TLR)2/6 ligand, UNE-C1, to human papilloma virus type 16 (HPV-16)-derived peptide antigen, E7, and found that the UNE-C1-conjugated cancer vaccine (UCV) showed significantly enhanced antitumor activity in vivo compared with the noncovalent combination of UNE-C1 and E7. The combination of UCV with PD-1 blockades further augmented its therapeutic efficacy. Specifically, the conjugation of UNE-C1 to E7 enhanced its retention in inguinal draining lymph nodes, the specific delivery to dendritic cells and E7 antigen-specific T cell responses, and antitumor efficacy in vivo compared with the noncovalent combination of the two peptides. These findings suggest the potential of UNE-C1 derived from human cysteinyl-tRNA synthetase 1 as a unique vehicle for the specific delivery of cancer antigens to antigen-presenting cells via TLR2/6 for the improvement of cancer vaccines.
Collapse
Affiliation(s)
- Hyeong Yun Kim
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Seongmin Cho
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Sang Bum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Republic of Korea
| | - Ee Chan Song
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Wonchul Jung
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Yun Gyeong Shin
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Ji Hun Suh
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Jihye Choi
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Ina Yoon
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Uijoo Kim
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Hamin Ban
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Sunkyo Hwang
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Jeongwon Mun
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Joohee Park
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Nayoung Kim
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea
| | - Youngjin Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Myung Hee Kim
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sunghoon Kim
- Institute for Artificial Intelligence and Biomedical Research (AIBI), Medicinal Bioconvergence Research Center, College of Pharmacy, Yonsei University, Incheon 21983, Republic of Korea; College of Medicine, Gangnam Severance Hospital, Yonsei University, Seoul 06273, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea; Interdisciplinary Graduate Program in Integrative Biotechnology & College of Medicine, Gangnam Severance Hospital, Yonsei University, Incheon 21983, Republic of Korea.
| |
Collapse
|
20
|
Foda BM, Misek SA, Gallo KA, Neubig RR. Inhibition of the Rho/MRTF pathway improves the response of BRAF-resistant melanoma to PD1/PDL1 blockade. Int J Cancer 2024; 155:1303-1315. [PMID: 38898604 DOI: 10.1002/ijc.35056] [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: 12/21/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024]
Abstract
Metastatic cutaneous melanoma is a fatal skin cancer. Resistance to targeted and immune therapies limits the benefits of current treatments. Identifying and adding anti-resistance agents to current treatment protocols can potentially improve clinical responses. Myocardin-related transcription factor (MRTF) is a transcriptional coactivator whose activity is indirectly regulated by actin and the Rho family of GTPases. We previously demonstrated that development of BRAF inhibitor (BRAFi) resistance frequently activates the Rho/MRTF pathway in human and mouse BRAFV600E melanomas. In clinical trials, pretreatment with BRAFi reduces the benefit of immune therapies. We aimed to test the efficacy of concurrent treatment with our MRTF pathway inhibitor CCG-257081 and anti-PD1 in vivo and to examine its effects on the melanoma immune microenvironment. Because MRTF pathway activation upregulates the expression of immune checkpoint inhibitor genes/proteins, we asked whether CCG-257081 can improve the response to immune checkpoint blockade. CCG-257081 reduced the expression of PDL1 in BRAFi-resistant melanoma cells and decreased surface PDL1 levels on both BRAFi-sensitive and -resistant melanoma cells. Using our recently described murine vemurafenib-resistant melanoma model, we found that CCG-257081, in combination with anti-PD1 immune therapy, reduced tumor growth and increased survival. Moreover, anti-PD1/CCG-257081 co-treatment increased infiltration of CD8+ T cells and B cells into the tumor microenvironment and reduced tumor-associated macrophages. Here, we propose CCG-257081 as an anti-resistance and immune therapy-enhancing anti-melanoma agent.
Collapse
Affiliation(s)
- Bardees M Foda
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
- Molecular Genetics and Enzymology Department, National Research Centre, Dokki, Egypt
| | - Sean A Misek
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, Michigan, USA
| | - Richard R Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
- Nicholas V. Perricone, M.D. Division of Dermatology, Department of Medicine, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
21
|
Singh KP, Singh A, Wolkenhauer O, Gupta SK. Regulatory Role of IL6 in Immune-Related Adverse Events during Checkpoint Inhibitor Treatment in Melanoma. Int J Mol Sci 2024; 25:10600. [PMID: 39408929 PMCID: PMC11476582 DOI: 10.3390/ijms251910600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The landscape of clinical management for metastatic melanoma (MM) and other solid tumors has been modernized by the advent of immune checkpoint inhibitors (ICI), including programmed cell death-1 (PD-1), programmed cell death-ligand 1 (PD-L1), and cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors. While these agents demonstrate efficacy in suppressing tumor growth, they also lead to immune-related adverse events (irAEs), resulting in the exacerbation of autoimmune diseases such as rheumatoid arthritis (RA), ulcerative colitis (UC), and Crohn's disease (CD). The immune checkpoint inhibitors offer promising advancements in the treatment of melanoma and other cancers, but they also present significant challenges related to irAEs and autoimmune diseases. Ongoing research is crucial to better understand these challenges and develop strategies for mitigating adverse effects while maximizing therapeutic benefits. In this manuscript, we addressed this challenge using network-based approaches by constructing and analyzing the molecular and signaling networks associated with tumor-immune crosstalk. Our analysis revealed that IL6 is the key regulator responsible for irAEs during ICI therapies. Furthermore, we conducted an integrative network and molecular-level analysis, including virtual screening, of drug libraries, such as the Collection of Open Natural Products (COCONUT) and the Zinc15 FDA-approved library, to identify potential IL6 inhibitors. Subsequently, the compound amprenavir was identified as the best molecule that may disrupt essential interactions between IL6 and IL6R, which are responsible for initiating the signaling cascades underlying irAEs in ICI therapies.
Collapse
Affiliation(s)
- Krishna P. Singh
- Department of Systems Biology & Bioinformatics, University of Rostock, 18051 Rostock, Germany; (K.P.S.); (O.W.)
| | - Anuj Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow 226028, India;
| | - Olaf Wolkenhauer
- Department of Systems Biology & Bioinformatics, University of Rostock, 18051 Rostock, Germany; (K.P.S.); (O.W.)
- Department of Biomedical Engineering & Bioinformatics, Chhattisgarh Swami Vivekananda Technical University, Bhilai 491107, India
- Leibniz Institute for Food Systems Biology, Technical University of Munich, 85354 Freising, Germany
| | - Shailendra Kumar Gupta
- Department of Systems Biology & Bioinformatics, University of Rostock, 18051 Rostock, Germany; (K.P.S.); (O.W.)
- Department of Biomedical Engineering & Bioinformatics, Chhattisgarh Swami Vivekananda Technical University, Bhilai 491107, India
| |
Collapse
|
22
|
Yildiz G, Torun Bayram M, Ünlü ŞM, Soylu A, Kavukçu S, Olgun N. Nivolumab-associated IgA Nephropathy in a Child With Malignant Melanoma. J Pediatr Hematol Oncol 2024; 46:e534-e536. [PMID: 39052862 DOI: 10.1097/mph.0000000000002931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/07/2024] [Indexed: 07/27/2024]
Abstract
Immune checkpoint inhibitors are humanized antibodies that inhibit downregulatory receptors on T cells, enhancing the antitumor activity of these cells. However, they have been associated with a wide range of systemic immune-related adverse events, including renal toxicities, among others. Most renal immune-related adverse events are acute interstitial nephritis causing acute kidney injury. Recently, immune checkpoint inhibitors-associated glomerular diseases, including IgA nephropathy, have been reported in adults. Most of the adult cases with glomerular involvement had also concomitant acute interstitial nephritis and acute kidney injury. We present the first pediatric case of IgA nephropathy without acute kidney injury during nivolumab treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Nur Olgun
- Department of Pediatric Oncology, Dokuz Eylul University Institute of Oncology, Izmir, Turkey
| |
Collapse
|
23
|
Ni R, Hu Z, Tao R. Advances of immune-checkpoint inhibition of CTLA-4 in pancreatic cancer. Biomed Pharmacother 2024; 179:117430. [PMID: 39260322 DOI: 10.1016/j.biopha.2024.117430] [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: 06/13/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024] Open
Abstract
Targeting checkpoints for immune cell activation has been acknowledged known as one of the most effective way to activate anti-tumor immune responses. Among them, drugs targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) are approved for clinical treatment though several more are in advanced stages of development, which demonstrated durable response rates and manageable safety profile. However, its therapy efficacy is unsatisfactory in pancreatic cancer (PC), which can be limited by the overall condition of patients, the pathological type of PC, the expression level of tumor related genes, etc. To improve clinical efficiency, various researches have been conducted, and the efficacy of combination therapy showed significantly improvement compared to monotherapy. This review analyzed current strategies based on anti-CTLA-4 combination immunotherapy, providing totally new idea for future research.
Collapse
Affiliation(s)
- Ran Ni
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhiming Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China; Department of Hepatobiliary & Pancreatic Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China.
| | - Ran Tao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
24
|
Xue F, Ren X, Kong C, Wang J, Liu L, Hu J, Shen N, Tang Z. Polymeric PD1/PDL1 bispecific antibody enhances immune checkpoint blockade therapy. Mater Today Bio 2024; 28:101239. [PMID: 39318373 PMCID: PMC11421358 DOI: 10.1016/j.mtbio.2024.101239] [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: 06/13/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Immune checkpoint blockade (ICB) therapy, particularly PD1/PDL1 inhibition, has demonstrated success in bolstering durable responses in patients. However, the response rate remains below 30 %. In this study, we developed a polymeric bispecific antibody (BsAb) targeting PD1/PDL1 to enhance ICB therapy. Specifically, poly(L-glutamic acid) (PGLU) was conjugated with a double cyclic Fc binding peptide, Fc-III-4C, through condensation reactions between the -COOH group of PGLU and the -NH2 group of Fc-III-4C. This conjugate was then mixed with αPD1 and αPDL1 monoclonal antibodies (mAbs) in an aqueous solution. Mechanistically, the PD1/PDL1 BsAb (BsAbαPD1+αPDL1) acts as a bridge between tumor cells and CD8+ T cells, continuously activating CD8+ T cells to a greater extent. This leads to significantly suppressed tumor growth and prolonged survival in a mouse model of colon cancer compared to treatment with either a single mAb or a mixture of free mAbs. The tumor suppression rate achieved by the BsAbαPD1+αPDL1 was 90.1 %, with a corresponding survival rate of 83.3 % after 48 days. Thus, this study underscores the effectiveness of the BsAbαPD1+αPDL1 as a synchronizing T cell engager and dual ICBs, offering theoretical guidance for clinical ICB therapy.
Collapse
Affiliation(s)
- Fuxin Xue
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Xitong Ren
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Chaoying Kong
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Jianfeng Wang
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Linlin Liu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Junli Hu
- Key Laboratory of UV-Emitting Materials and Technology, Northeast Normal University, Ministry of Education, Changchun, Jilin, 130024, China
| | - Na Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| |
Collapse
|
25
|
Gaudio G, Martino E, Pellizzari G, Cavallone M, Castellano G, Omar A, Katselashvili L, Trapani D, Curigliano G. Developing combination therapies with biologics in triple-negative breast cancer. Expert Opin Biol Ther 2024; 24:1075-1094. [PMID: 39360776 DOI: 10.1080/14712598.2024.2408756] [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: 07/04/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
INTRODUCTION Novel compounds have entered the triple-negative breast cancer (TNBC) treatment algorithm, namely immune checkpoints inhibitors (ICIs), PARP inhibitors and antibody-drug conjugates (ADCs). The optimization of treatment efficacy can be enhanced with the use of combination treatments, and the incorporation of novel compounds. In this review, we discuss the combination treatments under development for the treatment of TNBC. AREAS COVERED The development of new drugs occurring in recent years has boosted the research for novel combinations to target TNBC heterogeneity and improve outcomes. ICIs, ADCs, tyrosine kinase inhibitors (TKIs), and PARP inhibitors have emerged as leading players in this new landscape, while other compounds like novel intracellular pathways inhibitors or cancer vaccines are drawing more and more interest. The future of TNBC is outlined in combination approaches, and based on new cancer targets, including many chemotherapy-free treatments. EXPERT OPINION A large number of TNBC therapies have either proved clinically ineffective or weighted by unacceptable safety profiles. Others, however, have provided promising results and are currently in late-stage clinical trials, while a few have actually changed clinical practice in recent years. As novel, more and more selective drugs come up, combination strategies focusing the concept of synergy are fully warranted for the future.
Collapse
Affiliation(s)
- Gilda Gaudio
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Enzo Martino
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Gloria Pellizzari
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Matteo Cavallone
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Grazia Castellano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Abeid Omar
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Nuclear Medicine, Kenyatta University Teaching Referral and Research Hospital, Nairobi, Kenya
| | - Lika Katselashvili
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology, Caucasus Medical Centre, Tbilisi, Georgia
| | - Dario Trapani
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Haemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
26
|
Xu H, Yang Y, Yan Y, Li M, Wu S, Cao L, Chen W, Luo H, He Y. Safety and Efficacy of Rechallenge With Immune Checkpoint Inhibitors in Advanced Solid Tumor: A Systematic Review and Meta-Analysis. Cancer Med 2024; 13:e70324. [PMID: 39463070 PMCID: PMC11513547 DOI: 10.1002/cam4.70324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/21/2024] [Accepted: 09/28/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have drastically shifted the current landscape toward a wide variety of malignancies. However, ICIs are interrupted owing immune-related adverse events (irAEs), therapy completion, and disease progression. The risk-benefit of rechallenged ICIs remains inconclusive. Herein, a systematic review and meta-analysis were conducted to evaluate the safety and efficacy of ICI rechallenge in the treatment of advanced solid tumor. METHODS PubMed, Web of Science, Embase, and Cochrane Library were searched to analyze the efficacy and safety of ICI rechallenge. The study protocol was approved by the PROSPERO International Register of Systematic Reviews (CRD42022372222). The last updated search date was March 2, 2024. Objective response rate (ORR), disease control rate (DCR), overall survival (OS), and incidence rates of all- and high-grade irAEs were evaluated. RESULTS A total of 41 retrospective studies comprising 2343 patients were ultimately enrolled for qualitative and quantitative assessments. A total of 1200 (51.2%) individuals were male and the median age was 66 years (range 18-97 years). The majority of the tumors was lung cancer (n = 898, 38.3%). The occurrence rates of all-grade and high-grade (grade 3 or 4) irAEs between initial and readministration ICIs were not significantly different (all-grade: OR, 0.75, 95% CI: 0.39-1.45, p = 0.40; I2 = 87%; high-grade: OR, 0.96, 95% CI: 0.62-1.49, p = 0.87, I2 = 65%). ICIs restart presented a decreased ORR and DCR compared to initial ICI administration (ORR: OR, 0.36, 95% CI: 0.23-0.56, p < 0.00001; I2 = 67%; DCR: OR, 0.62, 95% CI: 0.43-0.89, p = 0.010; I2 = 53%). Seven studies with 513 patients for survival analysis revealed a nonsignificant difference in OS between the ICIs rechallenge and discontinuation cohorts (hazard ratio [HR]: 0.68, 95% confidence interval (CI): 0.35 to 1.35, p = 0.27). CONCLUSION Rechallenging immunotherapy is feasible, and patients should be carefully evaluated by a multidisciplinary team prior to initial therapy for close monitoring and assessment of the risk-benefit ratio. Therefore, prospective trials are essential to guide clinicians in the decision-making process. PROSPERO Registration: CRD42022372222.
Collapse
Affiliation(s)
- Huijun Xu
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Yang Yang
- Division of Life Sciences and Medicine, Department of Ultrasound, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Ying Yan
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Mengge Li
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Shusheng Wu
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Lulu Cao
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Wenju Chen
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Huiqin Luo
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| | - Yifu He
- Division of Life Sciences and Medicine, Department of Oncology, the First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiAnhuiChina
| |
Collapse
|
27
|
Morrissey S, Vasconcelos AG, Wang CL, Wang S, Cunha GM. Pooled Rate of Pseudoprogression, Patterns of Response, and Tumor Burden Analysis in Patients Undergoing Immunotherapy Oncologic Trials for Different Malignancies. Clin Oncol (R Coll Radiol) 2024; 36:624-631. [PMID: 38937187 DOI: 10.1016/j.clon.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
AIMS Assess rates of true pseudoprogression in unconfirmed progressive disease (iUPD) in a pool of immunotherapy clinical trials for different cancers, analyze tumor characteristics that drive iUPD classification, and investigate potentials predictors of pseudoprogression. MATERIALS AND METHODS Retrospective interpretation of prospectively acquired data. Patients from 18 immunotherapy clinical trials with two arms (RECIST 1.1, iRECIST), of 10 cancer types were selected. Pooled rate of true pseudoprogression among iUPD was estimated using a common effect meta-analysis. Target, Non-target, and new lesions as the trigger of confirmed-vs pseudo-progression were compared using Chi-Square and Fisher exact tests. Conditional logistic regression was used to investigate the association between age, sex, tumor burden at baseline, and number of follow ups and pseudoprogression. RESULTS 60/287 (21%) patients (17 women) were classified as iUPD with at least one subsequent confirmatory timepoint. The overall pooled estimate of pseudoprogression was 15% (95%CI: 8%--26%). Nontarget lesions were significantly more frequent the cause of iUPD than change in Target lesions size (p< 0.001). Most observations of true pseudoprogression occurred in the first follow-up (77%), whereas confirmed progression occurred in later time points during the trial. Pseudoprogression was not significantly associated with age, sex, tumor burden at baseline, or number of timepoints. CONCLUSION In a pool of immunotherapy trials, the rate of true pseudoprogression was 15%, most often in the first timepoint after baseline than later in treatment. iUPD categorization was mostly driven by changes in NT lesions rather than objective changes in measurements of target lesions.
Collapse
Affiliation(s)
- S Morrissey
- OncoRad Research Core, Department of Radiology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - A G Vasconcelos
- Department of Statistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - C L Wang
- Department of Radiology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - S Wang
- OncoRad Research Core, Department of Radiology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - G M Cunha
- OncoRad Research Core, Department of Radiology, Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA.
| |
Collapse
|
28
|
Bo Y, Wang H. Biomaterial-Based In Situ Cancer Vaccines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210452. [PMID: 36649567 PMCID: PMC10408245 DOI: 10.1002/adma.202210452] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Cancer immunotherapies have reshaped the paradigm for cancer treatment over the past decade. Among them, therapeutic cancer vaccines that aim to modulate antigen-presenting cells and subsequent T cell priming processes are among the first FDA-approved cancer immunotherapies. However, despite showing benign safety profiles and the capability to generate antigen-specific humoral and cellular responses, cancer vaccines have been limited by the modest therapeutic efficacy, especially for immunologically cold solid tumors. One key challenge lies in the identification of tumor-specific antigens, which involves a costly and lengthy process of tumor cell isolation, DNA/RNA extraction, sequencing, mutation analysis, epitope prediction, peptide synthesis, and antigen screening. To address these issues, in situ cancer vaccines have been actively pursued to generate endogenous antigens directly from tumors and utilize the generated tumor antigens to elicit potent cytotoxic T lymphocyte (CTL) response. Biomaterials-based in situ cancer vaccines, in particular, have achieved significant progress by taking advantage of biomaterials that can synergize antigens and adjuvants, troubleshoot delivery issues, home, and manipulate immune cells in situ. This review will provide an overview of biomaterials-based in situ cancer vaccines, either living or artificial materials, under development or in the clinic, and discuss the design criteria for in situ cancer vaccines.
Collapse
Affiliation(s)
- Yang Bo
- 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
- 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
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| |
Collapse
|
29
|
Deng M, Liu X, Jiang Y, Luo R, Xu L, Zhang X, Su J, Xu C, Hou Y. Tertiary lymphoid structures' pattern and prognostic value in primary adenocarcinoma of jejunum and ileum. World J Surg Oncol 2024; 22:261. [PMID: 39350287 PMCID: PMC11441114 DOI: 10.1186/s12957-024-03543-x] [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: 07/03/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024] Open
Abstract
To date, there have been no reports on tertiary lymphoid structures (TLS) in primary adenocarcinoma of jejunum and ileum. In this study, we employed digital pathology image analysis software to classify and quantify TLS, and evaluated the maturity of TLS using immunohistochemistry. Molecular genetics and immunotherapy biomarker detection were performed using next-generation sequencing technology, such as tumor mutational burden (TMB) and microsatellite instability (MSI). The aim of this study was to investigate the presence, location, maturity, association with immunotherapy biomarkers, and prognostic value of TLS in primary adenocarcinoma of jejunum and ileum. Compared to secondary follicle-like TLS (SFL-TLS), intra-tumoral TLS (IT-TLS) were more likely to manifest as early TLS (E-TLS) (P = 0.007). Compared to IT-TLS, SFL-TLS had a higher propensity to occur at the invasive margin (IM) (P = 0.032) and showed a trend towards being more prevalent at the tumor periphery (P = 0.057). In terms of immunotherapy biomarkers, there was a higher trend of IM-TLS density in PD-L1(22C3) score CPS < 1 group compared to PD-L1(22C3) score CPS ≥ 1 group (P = 0.071). TMB-H was significantly associated with MSI-H (P = 0.040). Univariate survival analysis demonstrated a correlation between high SFL-TLS group and prolonged disease free survival (DFS) (P = 0.047). There was also a trend towards prolonged DFS in the E-TLS-high group compared to the E-TLS-low group (P = 0.069). The peri-tumoral TLS (PT-TLS)-high group showed a trend of prolonged overall survival (OS) compared to the PT-TLS-low group (P = 0.090). In conclusion, the majority of TLS were located at the invasive margin and tumor periphery, predominantly consisting of mature TLS, while IT-TLS were mainly immature. Notably, TMB was closely associated with MSI and PD-L1, indicating potential predictive value for immunotherapy in primary adenocarcinoma of jejunum and ileum.
Collapse
Affiliation(s)
- Minying Deng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Liu
- Department of Pathology, Eye & ENT Hospital, Fudan University, Shanghai, 200032, China
| | - Yan Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xiaolei Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jieakesu Su
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
30
|
Hashemi M, Mohandesi Khosroshahi E, Tanha M, Khoushab S, Bizhanpour A, Azizi F, Mohammadzadeh M, Matinahmadi A, Khazaei Koohpar Z, Asadi S, Taheri H, Khorrami R, Ramezani Farani M, Rashidi M, Rezaei M, Fattah E, Taheriazam A, Entezari M. Targeting autophagy can synergize the efficacy of immune checkpoint inhibitors against therapeutic resistance: New promising strategy to reinvigorate cancer therapy. Heliyon 2024; 10:e37376. [PMID: 39309904 PMCID: PMC11415696 DOI: 10.1016/j.heliyon.2024.e37376] [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: 04/06/2024] [Revised: 06/29/2024] [Accepted: 09/02/2024] [Indexed: 09/25/2024] Open
Abstract
Immune checkpoints are a set of inhibitory and stimulatory molecules/mechanisms that affect the activity of immune cells to maintain the existing balance between pro- and anti-inflammatory signaling pathways and avoid the progression of autoimmune disorders. Tumor cells can employ these checkpoints to evade immune system. The discovery and development of immune checkpoint inhibitors (ICIs) was thereby a milestone in the area of immuno-oncology. ICIs stimulate anti-tumor immune responses primarily by disrupting co-inhibitory signaling mechanisms and accelerate immune-mediated killing of tumor cells. Despite the beneficial effects of ICIs, they sometimes encounter some degrees of therapeutic resistance, and thereby do not effectively act against tumors. Among multiple combination therapies have been introduced to date, targeting autophagy, as a cellular degradative process to remove expired organelles and subcellular constituents, has represented with potential capacities to overcome ICI-related therapy resistance. It has experimentally been illuminated that autophagy induction blocks the immune checkpoint molecules when administered in conjugation with ICIs, suggesting that autophagy activation may restrict therapeutic challenges that ICIs have encountered with. However, the autophagy flux can also provoke the immune escape of tumors, which must be considered. Since the conventional FDA-approved ICIs have designed and developed to target programmed cell death receptor/ligand 1 (PD-1/PD-L1) as well as cytotoxic T lymphocyte-associated molecule 4 (CTLA-4) immune checkpoint molecules, we aim to review the effects of autophagy targeting in combination with anti-PD-1/PD-L1- and anti-CTLA-4-based ICIs on cancer therapeutic resistance and tumor immune evasion.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahsa Tanha
- Department of Biological Sciences, University of Alabama, Tuscaloosa, AL, United States
| | - Saloomeh Khoushab
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Anahita Bizhanpour
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Farnaz Azizi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahsa Mohammadzadeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Arash Matinahmadi
- Department of Cellular and Molecular Biology, Nicolaus Copernicus University, Torun, Poland
| | - Zeinab Khazaei Koohpar
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hengameh Taheri
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Marzieh Ramezani Farani
- Department of Biological Sciences and Bioengineering, Nano Bio High-Tech Materials Research Center, Inha University, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Republic of Korea
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahdi Rezaei
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Eisa Fattah
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
31
|
Liu L, Zhou Z, Xie C, Hu L. Combination of bulk RNA and single-cell sequencing unveils PANoptosis-related immunological ecology hallmarks and classification for clinical decision-making in hepatocellular carcinoma. Sci Rep 2024; 14:22517. [PMID: 39342037 PMCID: PMC11438900 DOI: 10.1038/s41598-024-73847-1] [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: 04/20/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024] Open
Abstract
PANoptosis is engaged in the program of immune response and carcinogenicity. Nonetheless, the actual impacts of PANoptosis on clinical management and oncology immunity in hepatocellular carcinoma (HCC) are not fully grasped. RNA-seq-derived computations were conducted to sort out the molecular subtypes and elucidate the disparities based on PANoptosis molecules. Single-cell sequencing (scRNA-seq) tools including Cytotrace and Addmodulescore were extracted to characterize diversification potency and quantify the PANoptosis motion. Transcriptional factors were inferred by the pySCENIC package and Cellchat program scrutinized the intercellular exchange across cell compartments. The PANoptosis score system originated by incorporating 10 machine learning algorithms and 101 compositions to project clinical results and deteriorate tendencies. Circulatory PANoptosis-associated protein HSP90AA1 was determined by enzyme-linked immunosorbent assay (ELISA). HCC individuals could be categorized into low- and high-PANoptosis groups with diverse biogenic and pharmacotherapy heterogeneity. Individuals in the elevated PANoptosis subtype were characterized as "hot tumor" conveying the increased presence of immunogenicity while reiterating an explicit negative connection with tumor stemness. Compared to immune and stromal cells, cancerous cells showcased decreased PANoptosis and heightened PANoptosis malignant cell subgroups might be tied to a substantial level of genomic expression of SREBF2, JUND, GATAD1, ZBTB20, SMAD5 and implied a more aggressive potential. The PANoptosis index, derived from machine learning, has been established to provide succinct frameworks for predicting outcomes and clarified the noteworthy utility of conventional regimens, as the differentiated power of HCC occurred together with vascular invasion and hepatocellular adenoma (HCA). The experiment confirmed that the circulating HSP90AA1 was aberrantly augmented in HCC patients, thus demonstrating its potential as a discriminatory biomarker. We systematically deciphered the molecular and immune ecosystem traits of PANoptosis in bulk and scRNA-seq degrees, which may deliver advantageous insights for customized treatment, awareness of the pathological process and prognosis scrutiny for HCC patients.
Collapse
Affiliation(s)
- Li Liu
- Department of Pathology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhangxu Zhou
- Department of Clinical Laboratory, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| | - Cong Xie
- Department of Clinical Laboratory, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China.
| | - Liyi Hu
- Department of Clinical Laboratory, The People's Hospital of Chongqing Liang Jiang New Area, Chongqing, China.
| |
Collapse
|
32
|
Guo KC, Wang ZZ, Su XQ. Chinese Medicine in Colorectal Cancer Treatment: From Potential Targets and Mechanisms to Clinical Application. Chin J Integr Med 2024:10.1007/s11655-024-4115-8. [PMID: 39331211 DOI: 10.1007/s11655-024-4115-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 09/28/2024]
Abstract
Colorectal cancer (CRC) is a global health challenge necessitating innovative therapeutic strategies. There is an increasing trend toward the clinical application of integrative Chinese medicine (CM) and Western medicine approaches. Chinese herbal monomers and formulations exert enhanced antitumor effects by modulating multiple signaling pathways in tumor cells, including inhibiting cell proliferation, inducing apoptosis, suppressing angiogenesis, reversing multidrug resistance, inhibiting metastasis, and regulating immunity. The synergistic effects of CM with chemotherapy, targeted therapy, immunotherapy, and nanovectors provide a comprehensive framework for CRC treatment. CM can mitigate drug toxicity, improve immune function, control tumor progression, alleviate clinical symptoms, and improve patients' survival and quality of life. This review summarizes the key mechanisms and therapeutic strategies of CM in CRC, highlighting its clinical significance. The potential for CM and combination with conventional treatment modalities is emphasized, providing valuable insights for future research and clinical practice.
Collapse
Affiliation(s)
- Ke-Chen Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zao-Zao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xiang-Qian Su
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
33
|
Bhat AA, Afzal M, Moglad E, Thapa R, Ali H, Almalki WH, Kazmi I, Alzarea SI, Gupta G, Subramaniyan V. lncRNAs as prognostic markers and therapeutic targets in cuproptosis-mediated cancer. Clin Exp Med 2024; 24:226. [PMID: 39325172 PMCID: PMC11427524 DOI: 10.1007/s10238-024-01491-0] [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: 08/11/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Long non-coding RNAs (lncRNAs) have emerged as crucial regulators in various cellular processes, including cancer progression and stress response. Recent studies have demonstrated that copper accumulation induces a unique form of cell death known as cuproptosis, with lncRNAs playing a key role in regulating cuproptosis-associated pathways. These lncRNAs may trigger cell-specific responses to copper stress, presenting new opportunities as prognostic markers and therapeutic targets. This paper delves into the role of lncRNAs in cuproptosis-mediated cancer, underscoring their potential as biomarkers and targets for innovative therapeutic strategies. A thorough review of scientific literature was conducted, utilizing databases such as PubMed, Google Scholar, and ScienceDirect, with search terms like 'lncRNAs,' 'cuproptosis,' and 'cancer.' Studies were selected based on their relevance to lncRNA regulation of cuproptosis pathways and their implications for cancer prognosis and treatment. The review highlights the significant contribution of lncRNAs in regulating cuproptosis-related genes and pathways, impacting copper metabolism, mitochondrial stress responses, and apoptotic signaling. Specific lncRNAs are potential prognostic markers in breast, lung, liver, ovarian, pancreatic, and gastric cancers. The objective of this article is to explore the role of lncRNAs as potential prognostic markers and therapeutic targets in cancers mediated by cuproptosis.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al Kharj, Saudi Arabia
| | - Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - 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
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor Darul Ehsan, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500, Subang Jaya, Selangor, Malaysia.
| |
Collapse
|
34
|
Passang T, Wang S, Zhang H, Zeng F, Hsu PC, Wang W, Li JM, Liu Y, Ravindranathan S, Lesinski GB, Waller EK. VPAC2 Receptor Signaling Promotes Growth and Immunosuppression in Pancreatic Cancer. Cancer Res 2024; 84:2954-2967. [PMID: 38809694 PMCID: PMC11458156 DOI: 10.1158/0008-5472.can-23-3628] [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: 11/17/2023] [Revised: 03/29/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) harbors a complex tumor microenvironment, and cross-talk among cells in the tumor microenvironment can contribute to drug resistance and relapse. Vasoactive intestinal peptide (VIP) is overexpressed in PDAC, and VIP receptors expressed on T cells are a targetable pathway that sensitizes PDAC to immunotherapy. In this study, we showed that pancreatic cancer cells engage in autocrine VIP signaling through VIP receptor 2 (VPAC2). High coexpression of VIP with VPAC2 correlated with reduced relapse-free survival in patients with PDAC. VPAC2 activation in PDAC cells upregulated Piwi-like RNA-mediated gene silencing 2, which stimulated cancer cell clonogenic growth. In addition, VPAC2 signaling increased expression of TGFβ1 to inhibit T-cell function. Loss of VPAC2 on PDAC cells led to reduced tumor growth and increased sensitivity to anti-PD-1 immunotherapy in mouse models of PDAC. Overall, these findings expand our understanding of the role of VIP/VPAC2 signaling in PDAC and provide the rationale for developing potent VPAC2-specific antagonists for treating patients with PDAC. Significance: Autocrine VIP signaling via VPAC2 promotes cancer cell growth and inhibits T-cell function in pancreatic ductal adenocarcinoma, highlighting its potential as a therapeutic target to improve pancreatic cancer treatment.
Collapse
Affiliation(s)
- Tenzin Passang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Shuhua Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Hanwen Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Fanyuan Zeng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Po-Chih Hsu
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Wenxi Wang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jian Ming Li
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Yuan Liu
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Sruthi Ravindranathan
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Gregory B. Lesinski
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| | - Edmund K. Waller
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Winship Cancer Institute Emory University, Atlanta, GA, USA
| |
Collapse
|
35
|
Kim DH, Ahn JS, Kang M, Park G, Lim Y, Hwang S, Ock CY, Koh J, Chung EJ, Kwon SK, Jeon YK, Jung KC, Ahn SH, Keam B. Comparison of Tumor Microenvironments between Primary Tumors and Lymph Node Metastases in Head and Neck Squamous Cell Carcinoma and Their Predictive Role in Immune Checkpoint Inhibitor Treatment. Cells 2024; 13:1557. [PMID: 39329741 PMCID: PMC11429639 DOI: 10.3390/cells13181557] [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: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
The relationship between tumor microenvironments (TMEs) of regional lymph node metastases (LNMs) and primary tumors in head and neck squamous cell carcinoma (HNSCC) remains unclear. This study compared tumor-infiltrating lymphocytes (TILs) and the immune phenotype (IP), characterized by spatial TIL distribution, between primary tumors and LNMs. Twenty-one HNSCC patients with regional LNM who received immune checkpoint inhibitors (ICIs) were included. A paired comparative analysis of TIL densities and IP between primary tumors and LNMs revealed no significant difference or correlation between TIL densities in primary tumors and LNMs. Their IPs were discordant in 12 patients (57.1%). Patients with high intratumoral TIL exhibited longer progression-free survival (PFS) than those with low intratumoral TIL in both primary tumors (median, 5.2 vs. 1.3 months, p = 0.003) and LNMs (median, 30.2 vs. 1.3 months, p = 0.012). Patients with inflamed IP exhibited longer PFS than those with non-inflamed IP in both primary tumors (median, 4.5 vs. 1.3 months, p = 0.043) and LNMs (median, 4.1 vs. 1.3 months, p = 0.037). Given the lack of correlation in TIL densities, the discrepancies in IP, and the predictive value of both TMEs, evaluating the TMEs of both primary tumors and LNMs may be beneficial for the precise use of ICIs in HNSCC. There was a significant discordance between the TME of primary tumors and LNMs, with implications in survival outcomes. Therefore, evaluating the TME of both the primary tumor and LNM could be beneficial for the precise use of ICIs in HNSCC.
Collapse
Affiliation(s)
- Dong Hyun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | | | - Mingu Kang
- Lunit Inc., Seoul 06241, Republic of Korea
| | - Gahee Park
- Lunit Inc., Seoul 06241, Republic of Korea
| | - Yoojoo Lim
- Lunit Inc., Seoul 06241, Republic of Korea
| | | | | | - Jiwon Koh
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Eun-Jae Chung
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Seong-Keun Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Yoon Kyung Jeon
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Kyeong Cheon Jung
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Soon-Hyun Ahn
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| |
Collapse
|
36
|
Nakatsukasa H, Takahashi M, Shibano M, Ishigami Y, Kawaguchi T, Nakamura Y, Kaneda H. Clinical impact of concomitant BIO-three use in advanced or recurrent non-small cell lung cancer treated with immune-checkpoint inhibitor. Int J Clin Oncol 2024:10.1007/s10147-024-02622-z. [PMID: 39278980 DOI: 10.1007/s10147-024-02622-z] [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: 04/30/2024] [Accepted: 09/01/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been approved as first-line therapy for advanced non-small cell lung cancer (NSCLC). The probiotic MIYAIRI 588 can potentially improve the outcomes of patients with advanced NSCLC treated with ICI. However, the impact of other probiotics on ICI-treatment efficacy remains unclear. Thus, we aimed to clarify the association between BIO-three use and treatment outcomes in patients with advanced NSCLC treated with ICI. METHODS This retrospective study included patients aged ≥ 18 years with advanced or recurrent NSCLC who had received ICI monotherapy or ICI plus chemotherapy. Concomitant therapy with probiotic bacteria was defined as receiving it within 180 days before ICI therapy. RESULTS Here, 289 patients were enrolled, including 23 (8.0%) receiving BIO-three. In the multivariable analysis, the progression-free survival (PFS) and overall survival (OS) of patients receiving BIO-three tended to be longer than those of patients not receiving probiotic therapy (PFS, hazard ratio [HR] 0.75; 95% confidence interval [CI] 0.43-1.30; p = 0.33; OS, HR 0.69; 95% CI 0.37-1.28; p = 0.24). After propensity score matching with weighted adjustment, patients receiving BIO-three tended to have prolonged PFS (median PFS [range] 7.6 months [2.6-17.4] vs 3.2 months [1.6-7.0]; HR 0.53; 95% CI 0.25-1.12; p = 0.09) and OS (median OS [range] 25.6 months [10.8-not reached] vs 10.9 months [7.3-not reached]; HR 0.57; 95% CI 0.24-1.36; p = 0.20) than those not receiving probiotic therapy. CONCLUSION This study suggests the prognostic impact of concomitant BIO-three use in patients with advanced NSCLC on ICI treatment.
Collapse
Affiliation(s)
- Hitomi Nakatsukasa
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masaya Takahashi
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
- Department of Quality and Safety Management, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Masahito Shibano
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Yusuke Ishigami
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Yasutaka Nakamura
- Department of Pharmacy, Osaka Metropolitan University Hospital, Osaka, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan.
| |
Collapse
|
37
|
Lustenberger SK, Castro Jaramillo CA, Bärtschi LA, Williams R, Schibli R, Mu L, Krämer SD. Towards imaging the immune state of cancer by PET: Targeting legumain with 11C-labeled P1-Asn peptidomimetics carrying a cyano-warhead. Nucl Med Biol 2024; 138-139:108951. [PMID: 39303441 DOI: 10.1016/j.nucmedbio.2024.108951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
PURPOSE M2-type tumor-associated macrophages (TAM) residing in the tumor microenvironment (TME) have been linked to tumor invasiveness, metastasis and poor prognosis. M2 TAMs suppress T cell activation, silencing the recognition of the cancer by the immune system. Targeting TAMs in anti-cancer therapy may support the immune system and immune-checkpoint inhibitor therapies to fight the cancer cells. We aimed to develop a PET tracer for the imaging of M2 TAM infiltration of cancer, using activated legumain as the imaging target. BASIC PROCEDURES Two P1-mimicking inhibitors with a cyano-warhead were labeled with carbon-11 and evaluated in vitro and in vivo with a CT26 tumor mouse model. Target expression and activity were quantified from RT-qPCR and in vitro substrate conversion, respectively. The co-localization of legumain and TAMs was assessed by fluorescence microscopy. The two tracers were evaluated by PET with subsequent biodistribution analysis with the dissected tissues. Parent-to-total radioactivity in plasma was determined at several time points after i.v. tracer injection, using reverse phase radio-UPLC. MAIN FINDINGS Legumain displayed a target density of 40.7 ± 19.1 pmol per mg total protein in tumor lysate (n = 4) with high substrate conversion and colocalization with M2 macrophages in the tumor periphery. [11C]1 and [11C]2 were synthesized with >95 % radiochemical purity and 12.9-382.2 GBq/μmol molar activity at the end of synthesis. We observed heterogeneous tumor accumulation in in vitro autoradiography and PET for both tracers. However, excess unlabeled 1 or 2 did not compete with tracer accumulation. Both [11C]1 and [11C]2 were rapidly metabolized to a polar radiometabolite in vivo. PRINCIPAL CONCLUSIONS The legumain tracers [11C]1 and [11C]2, synthesized with high radiochemical purity and molar activity, accumulate in the legumain-positive CT26 tumor in vivo. However, the lack of competition by excess compound questions their specificity. Both tracers are rapidly metabolized in vivo, requiring structural modifications towards more stable tracers for further investigations.
Collapse
Affiliation(s)
- Severin K Lustenberger
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Claudia A Castro Jaramillo
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Lena A Bärtschi
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Rich Williams
- Queens University Belfast, BT7 1NN Belfast, United Kingdom of Great Britain and Northern Ireland
| | - Roger Schibli
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Linjing Mu
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Stefanie D Krämer
- Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, ETH Zürich, 8092 Zürich, Switzerland.
| |
Collapse
|
38
|
Patel AM, Willingham A, Cheng AC, Tomazela D, Bowman E, Kofman E, Zhang F, Bao J, Sanzone JR, Choy JW, Flygare JA, Han JH, Pradhan K, Kieffer M, Chernyak N, Akbari P, Liu P, Mehmood R, Naravula S, Hollingsworth SA, Bhagwat B, Lang SB, Seganish WM. Design and Optimization of Selectivity-Tunable Toll-like Receptor 7/8 Agonists as Novel Antibody-Drug Conjugate Payloads. J Med Chem 2024; 67:15756-15779. [PMID: 39172064 DOI: 10.1021/acs.jmedchem.4c01384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Toll-like receptors 7 and 8 are involved in modulating the adaptive and innate immune responses, and their activation has shown promise as a therapeutic strategy in the field of immuno-oncology. While systemic exposure to TLR7/8 agonists can result in poor tolerance, combination therapies and targeted delivery through antibody-drug conjugates (ADCs) can help mitigate adverse effects. Described herein is the identification of a novel and potent series of pyrazolopyrimidine-based TLR7/8 agonists with tunable receptor selectivity. Representative agonists from this series were successfully able to induce the production of various proinflammatory cytokines and chemokines from human peripheral blood mononuclear cells. Anti-HER2-25 and anti-HER2-26 ADCs made from this class of payloads demonstrated mechanism-based activation of TLR7/8 in a THP1/N87 coculture system.
Collapse
Affiliation(s)
- Akash M Patel
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Aarron Willingham
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Alan C Cheng
- Modeling and Informatics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Daniela Tomazela
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Eddie Bowman
- Discovery Oncology, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Esther Kofman
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Fan Zhang
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Jianming Bao
- External Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Jillian R Sanzone
- External Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Jonathan W Choy
- Discovery Oncology, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - John A Flygare
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Jin-Hwan Han
- Discovery Oncology, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Komal Pradhan
- Discovery Oncology, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Madeleine Kieffer
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Natalia Chernyak
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Peyman Akbari
- Discovery Oncology, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Ping Liu
- External Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Rimsha Mehmood
- Modeling and Informatics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Saraswathi Naravula
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Scott A Hollingsworth
- Modeling and Informatics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Bhagyashree Bhagwat
- Discovery Biologics, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - Simon B Lang
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| | - W Michael Seganish
- Discovery Chemistry, Merck & Co. Inc., South San Francisco, California 94080, United States
| |
Collapse
|
39
|
Alkhawaja B, Abuarqoub D, Al-natour M, Alshaer W, Abdallah Q, Esawi E, Jaber M, Alkhawaja N, Ghanim BY, Qinna N, Watts AG. Facile Rebridging Conjugation Approach to Attain Monoclonal Antibody-Targeted Nanoparticles with Enhanced Antigen Binding and Payload Delivery. Bioconjug Chem 2024; 35. [PMID: 39254438 PMCID: PMC11487529 DOI: 10.1021/acs.bioconjchem.4c00275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
Adopting conventional conjugation approaches to construct antibody-targeted nanoparticles (NPs) has demonstrated suboptimal control over the binding orientation and the structural stability of monoclonal antibodies (mAbs). Hitherto, the developed antibody-targeted NPs have shown proof of concept but lack product homogeneity, batch-to-batch reproducibility, and stability, precluding their advancement toward the clinic. To circumvent these limitations and advance toward clinical application, herein, a refined approach based on site-specific construction of mAb-immobilized NPs will be appraised. Initially, the conjugation of atezolizumab (anti-PDL1 antibody, Amab) with polymeric NPs was developed using bis-haloacetamide (BisHalide) rebridging chemistry, followed by click chemistry (NP-Fab BisHalide Ab and NP-Fc BisHalide Ab). For comparison purposes, mAb-immobilized NPs developed utilizing conventional conjugation methods, namely, N-hydroxysuccinimide (NHS) coupling and maleimide chemistry (NP-NHS Ab and NP-Mal Ab), were included. Next, flow cytometry and confocal microscopy experiments evaluated the actively targeted NPs (loaded with fluorescent dye) for cellular binding and uptake. Our results demonstrated the superior and selective binding and uptake of NP-Fab BisHalide Ab and NP-Fc BisHalide Ab into EMT6 cells by 19-fold and 13-fold, respectively. To evaluate the PDL1-dependent cell uptake and the selectivity of the treatments, a blocking step of the PDL1 receptor with Amab was performed prior to incubation with NP-Fab BisHalide Ab and NP-Fc BisHalide Ab. To our delight, the binding and uptake of fluorescent NPs were reduced significantly by 3-fold for NP-Fab BisHalide Ab, demonstrating the PDL1-mediated uptake. Moreover, NP-Fab BisHalide Ab and NP-Fc BisHalide Ab were entrapped with the paclitaxel payload, and their cytotoxicity was evaluated. They showed significant enhancements compared to free paclitaxel and NP-NHS Ab. Overall, this work will provide a facile conjugation method that could be implemented to actively target NPs with a plethora of therapeutic mAbs approved for various malignancies.
Collapse
Affiliation(s)
- Bayan Alkhawaja
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
- Department
of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, U.K.
| | - Duaa Abuarqoub
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
- Cell
Therapy Center, University of Jordan, Amman 11942, Jordan
| | - Mohammad Al-natour
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
| | - Walhan Alshaer
- Cell
Therapy Center, University of Jordan, Amman 11942, Jordan
| | - Qasem Abdallah
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
| | - Ezaldeen Esawi
- Cell
Therapy Center, University of Jordan, Amman 11942, Jordan
| | - Malak Jaber
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
| | - Nour Alkhawaja
- Faculty
of Pharmacy and Medical Sciences, University
of Petra, Amman 11196, Jordan
| | - Bayan Y. Ghanim
- University
of Petra Pharmaceutical Center, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan
| | - Nidal Qinna
- University
of Petra Pharmaceutical Center, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan
| | - Andrew G. Watts
- Department
of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, U.K.
| |
Collapse
|
40
|
Zhou Q, Xu J, Chen X, Ouyang J, Mao C, Zhang Z. CD276 as a promising diagnostic and prognostic biomarker for bladder cancer through bioinformatics and clinical research. Front Oncol 2024; 14:1445526. [PMID: 39319055 PMCID: PMC11419956 DOI: 10.3389/fonc.2024.1445526] [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: 06/07/2024] [Accepted: 08/19/2024] [Indexed: 09/26/2024] Open
Abstract
Objective To assess CD276 expression and explore its relationship with the clinicopathological characteristics and prognosis of patients with bladder cancer. Methods In total, RNA-sequencing data and clinical profiles of 436 bladder cancer cases from The Cancer Genome Atlas (TCGA) were assessed using the University of California Santa Cruz Xena (UCSC) platform. We compared the CD276 levels in cancerous and adjacent normal tissues and used the R software for statistical association with the clinical stage, grade, and survival (the overall survival, disease-specific survival, and progression-free survival). A single-gene GSEA analysis on TCGA-BLCA data was performed to explore potential pathways through which CD276 might influence bladder cancer. Additionally, CD276 expression was analyzed by comparing data from 9 cancerous tissues and 3 adjacent normal tissues in the GEO dataset GSE7476. Furthermore, we analyzed 133 cancerous bladder and adjacent tissue samples from the Soochow University Hospital, collected between January 1, 2016, and September 30, 2022, to assess the CD276 protein expression using immunohistochemistry. We examined the relationship between tumor CD276 levels and clinical outcomes and prognosis of bladder cancer. Results Bioinformatic analysis revealed elevated CD276 expression in tumors compared to that in adjacent tissues (p<0.05), correlating with poor survival. GSEA revealed that CD276 was significantly involved in extracellular matrix-related pathways. Immunohistochemistry confirmed CD276 overexpression in tumor tissues, with higher levels linked to advanced pathological grades and worse prognosis. Conclusion CD276 is markedly upregulated in bladder cancer and associated with severe pathological features, advanced disease, potential for metastasis, and diminished survival rates. It may promote bladder cancer development and progression by influencing extracellular matrix-related-related pathways, making it a viable diagnostic and prognostic biomarker for bladder cancer.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianhao Xu
- Department of Pathology, The First People’s Hospital of Kunshan, Suzhou, China
| | - Xuelei Chen
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ouyang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caiping Mao
- Department of Reproductive Medicine Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhiyu Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
41
|
Ahmad W, Sajjad W, Zhou Q, Ge Z. Nanomedicine for combination of chemodynamic therapy and immunotherapy of cancers. Biomater Sci 2024; 12:4607-4629. [PMID: 39115141 DOI: 10.1039/d3bm02133e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Chemodynamic therapy (CDT), as a new type of therapy, has received more and more attention in the field of tumor therapy in recent years. By virtue of the characteristics of weak acidity and excess H2O2 in the tumor microenvironment, CDT uses the Fenton or Fenton-like reactions to catalyze the transformation of H2O2 into strongly oxidizing ˙OH, resulting in increased intracellular oxidative stress for lipid oxidation, protein inactivation, or DNA damage, and finally inducing apoptosis of cancer cells. In particular, CDT has the advantage of tumor specificity. However, the therapeutic efficacy of CDT frequently depends on the catalytic efficiency of the Fenton reaction, which needs the presence of sufficient H2O2 and catalytic metal ions. Relatively low concentrations of H2O2 and the lack of catalytic metal ions usually limit the final therapeutic effect. The combination of CDT with immunotherapy will be an effective means to improve the therapeutic effect. In this review paper, the recent progress related to nanomedicine for the combination of CDT and immunotherapy is summarized. Immunogenic death of tumor cells, immune checkpoint inhibitors, and stimulator of interferon gene (STING) activation as the main immunotherapy strategies to combine with CDT are discussed. Finally, the challenges and prospects for the clinical translation and future development direction are discussed.
Collapse
Affiliation(s)
- Waqas Ahmad
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Wasim Sajjad
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Zhishen Ge
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
42
|
He YL, Liu JY, Almgrami RT, Fan YZ, Zhang Y. Cancer immunotherapy of Wilms tumor: a narrative review. Future Oncol 2024; 20:2293-2302. [PMID: 39235074 PMCID: PMC11508995 DOI: 10.1080/14796694.2024.2386929] [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: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Wilms tumor (WT) is the most common malignant tumor of the urinary system in children. Though the traditional treatment of surgery plus radiotherapy and chemotherapy achieves exciting clinical efficacy, in relapsed and refractory cases, the long-term overall survival rates are poor. Besides, chemotherapy and radiation have serious long-term toxic side effects on children. Cancer immunotherapy is a new tumor therapy that works by activating the body's immune system to allow immune cells to kill tumor cells more efficiently. Currently, cancer immunotherapy has been tested in clinical trials or basic studies in WT. This article reviews the current status of clinical trials and basic research of cancer immunotherapy in WT to promote the application of cancer immunotherapy in WT patients.
Collapse
Affiliation(s)
- Yu Lin He
- Second Ward of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Yan Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rahma Taher Almgrami
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Zhong Fan
- Second Ward of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
43
|
Cao W, Xie Y, Cai L, Wang M, Chen Z, Wang Z, Xv J, Wang Y, Li R, Liu X, Wang W. Pan‑cancer analysis on the role of KMT2C expression in tumor progression and immunotherapy. Oncol Lett 2024; 28:444. [PMID: 39091583 PMCID: PMC11292467 DOI: 10.3892/ol.2024.14577] [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: 12/04/2023] [Accepted: 05/10/2024] [Indexed: 08/04/2024] Open
Abstract
Histone lysine N-methyltransferase 2C (KMT2C) is involved in transcriptional regulation and DNA damage repair. Mutations in KMT2C have been implicated in the progression, metastasis, and drug resistance of multiple cancer types. However, the roles of KMT2C in the regulation of tumor prognosis, immune cell infiltration and the immune microenvironment in these multiple cancer types remain unclear. Therefore, in the present study, data from The Cancer Genome Atlas and Genotype-Tissue Expression databases were used for KMT2C expression analyses. Kaplan-Meier and univariate Cox regression analyses were also performed to investigate the prognostic role of KMT2C. In addition, Gene Set Enrichment Analysis (GSEA) was conducted to study the KMT2C-related signaling pathways. Tumor immune estimation resource 2 and single-sample GSEA were conducted to investigate the correlation between KMT2C expression and immune cell infiltrations, and Spearman's analysis was conducted to study the correlations among KMT2C, tumor mutational burden, microsatellite instability, immune regulators, chemokines and immune receptors. Immunohistochemistry of patient kidney tumor samples was performed to verify the correlation between KMT2C and programmed death-ligand 1 (PD-L1) expression. Finally, RNA interference, wound healing and colony formation assays were conducted to evaluate the effects of KMT2C expression on cell proliferation and metastasis. The results of the present study demonstrated that KMT2C was highly expressed in multiple cancer types, was a protective factor in kidney renal clear cell carcinoma and ovarian serous cystadenocarcinoma, and a risk factor for lung squamous cell carcinoma and uveal melanoma. In addition, KMT2C levels were negatively correlated with immune-activated pathways and the infiltration of immune cells, and positively correlated with inhibitory immune factors and tumor angiogenesis. Patients with low KMT2C expression had higher objective response rates to immunotherapy, and drug sensitivity analysis indicated that topoisomerase, histone deacetylase, DOT1-like histone H3K79 methyltransferase and G9A nuclear histone lysine methyltransferase inhibitors could potentially be used to treat tumors with high KMT2C expression levels. Finally, the KMT2C and PD-L1 expression levels were shown to be positively correlated, and KMT2C knockdown markedly promoted the proliferation and invasion capacities of A549 cells. In conclusion, the present study revealed that low KMT2C expression may be a promising biomarker for predicting the response of patients with cancer to immunotherapy. Conversely, high KMT2C expression was shown to promote tumor angiogenesis, which may contribute to the formation of the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Wei Cao
- Department of Thoracic Surgery, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yawen Xie
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Li Cai
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
- Department of Pathology, School of Basic Medicine, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Mengqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Zhuoying Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Ziteng Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jiajia Xv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yuqing Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Rong Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xuesong Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wenliang Wang
- Institute of Clinical Immunology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
44
|
Tiersma JF, Evers B, Bakker BM, Reijngoud DJ, de Bruyn M, de Jong S, Jalving M. Targeting tumour metabolism in melanoma to enhance response to immune checkpoint inhibition: A balancing act. Cancer Treat Rev 2024; 129:102802. [PMID: 39029155 DOI: 10.1016/j.ctrv.2024.102802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024]
Abstract
Immune checkpoint inhibition has transformed the treatment landscape of advanced melanoma and long-term survival of patients is now possible. However, at least half of the patients do not benefit sufficiently. Metabolic reprogramming is a hallmark of cancer cells and may contribute to both tumour growth and immune evasion by the tumour. Preclinical studies have indeed demonstrated that modulating tumour metabolism can reduce tumour growth while improving the functionality of immune cells. Since metabolic pathways are commonly shared between immune and tumour cells, it is essential to understand how modulating tumour metabolism in patients influences the intricate balance of pro-and anti-tumour immune effects in the tumour microenvironment. The key question is whether modulating tumour metabolism can inhibit tumour cell growth as well as facilitate an anti-tumour immune response. Here, we review current knowledge on the effect of tumour metabolism on the immune response in melanoma. We summarise metabolic pathways in melanoma and non-cancerous cells in the tumour microenvironment and discuss models and techniques available to study the metabolic-immune interaction. Finally, we discuss clinical use of these techniques to improve our understanding of how metabolic interventions can tip the balance towards a favourable, immune permissive microenvironment in melanoma patients.
Collapse
Affiliation(s)
- J F Tiersma
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B Evers
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - D J Reijngoud
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M de Bruyn
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
45
|
Liu Y, Li N, Guo Y, Zhou Q, Yang Y, Lu J, Tian Z, Zhou J, Yan S, Li X, Shi L, Jiang S, Ge J, Feng R, Huang D, Zeng Z, Fan S, Xiong W, Li G, Zhang W. APLNR inhibited nasopharyngeal carcinoma growth and immune escape by downregulating PD-L1. Int Immunopharmacol 2024; 137:112523. [PMID: 38909500 DOI: 10.1016/j.intimp.2024.112523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND APLNR is a G protein-coupled receptor and our previous study had revealed that APLNR could inhibit nasopharyngeal carcinoma (NPC) growth and metastasis. However, the role of APLNR in regulating PD-L1 expression and immune escape in NPC is unknown. METHODS We analyzed the expression and correlation of APLNR and PD-L1 in NPC tissues and cells. We investigated the effect of APLNR on PD-L1 expression and the underlying mechanism in vitro and in vivo. We also evaluated the therapeutic potential of targeting APLNR in combination with PD-L1 antibody in a nude mouse xenograft model. RESULTS We found that APLNR was negatively correlated with PD-L1 in NPC tissues and cells. APLNR could inhibit PD-L1 expression by binding to the FERM domain of JAK1 and blocking the interaction between JAK1 and IFNGR1, thus suppressing IFN-γ-mediated activation of the JAK1/STAT1 pathway. APLNR could also inhibit NPC immune escape by enhancing IFN-γ secretion and CD8+ T-cell infiltration and reducing CD8+ T-cell apoptosis and dysfunction. Moreover, the best effect was achieved in inhibiting NPC growth in nude mice when APLNR combined with PD-L1 antibody. CONCLUSIONS Our study revealed a novel mechanism of APLNR regulating PD-L1 expression and immune escape in NPC and suggested that APLNR maybe a potential therapeutic target for NPC immunotherapy.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Nan Li
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yilin Guo
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Zhou
- Department of Clinical Laboratory, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Yuqin Yang
- Shenzhen Maternity &Child Healthcare Hospital Clinical Laboratory, Shenzhen, Guangdong, China
| | - Jiaxue Lu
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ziying Tian
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jieyu Zhou
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shiqi Yan
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Shi
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Su Jiang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Junshang Ge
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Ranran Feng
- Department of Andrology, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, China
| | - Donghai Huang
- Department of Otolaryngology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Medical Laboratory Science, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
46
|
Han ZY, Fu ZJ, Wang YZ, Zhang C, Chen QW, An JX, Zhang XZ. Probiotics functionalized with a gallium-polyphenol network modulate the intratumor microbiota and promote anti-tumor immune responses in pancreatic cancer. Nat Commun 2024; 15:7096. [PMID: 39154092 PMCID: PMC11330462 DOI: 10.1038/s41467-024-51534-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 08/12/2024] [Indexed: 08/19/2024] Open
Abstract
The intratumor microbiome imbalance in pancreatic cancer promotes a tolerogenic immune response and triggers immunotherapy resistance. Here we show that Lactobacillus rhamnosus GG probiotics, outfitted with a gallium-polyphenol network (LGG@Ga-poly), bolster immunotherapy in pancreatic cancer by modulating microbiota-immune interactions. Upon oral administration, LGG@Ga-poly targets pancreatic tumors specifically, and selectively eradicates tumor-promoting Proteobacteria and microbiota-derived lipopolysaccharides through a gallium-facilitated disruption of bacterial iron respiration. This elimination of intratumor microbiota impedes the activation of tumoral Toll-like receptors, thus reducing immunosuppressive PD-L1 and interleukin-1β expression by tumor cells, diminishing immunotolerant myeloid populations, and improving the infiltration of cytotoxic T lymphocytes in tumors. Moreover, LGG@Ga-poly hampers pancreatic tumor growth in both preventive and therapeutic contexts, and amplifies the antitumor efficacy of immune checkpoint blockade in preclinical cancer models in female mice. Overall, we offer evidence that thoughtfully designed biomaterials targeting intratumor microbiota can efficaciously augment immunotherapy for the challenging pancreatic cancer.
Collapse
Affiliation(s)
- Zi-Yi Han
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Zhuang-Jiong Fu
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Yu-Zhang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Cheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Jia-Xin An
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China.
| |
Collapse
|
47
|
Motevasseli M, Darvishi M, Khoshnevisan A, Zeinalizadeh M, Saffar H, Bayat S, Najafi A, Abbaspour MJ, Mamivand A, Olson SB, Tabrizi M. Distinct tumor-TAM interactions in IDH-stratified glioma microenvironments unveiled by single-cell and spatial transcriptomics. Acta Neuropathol Commun 2024; 12:133. [PMID: 39148129 PMCID: PMC11328419 DOI: 10.1186/s40478-024-01837-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 07/02/2024] [Indexed: 08/17/2024] Open
Abstract
Tumor-associated macrophages (TAMs) residing in the tumor microenvironment (TME) are characterized by their pivotal roles in tumor progression, antitumor immunity, and TME remodeling. However, a thorough comparative characterization of tumor-TAM crosstalk across IDH-defined categories of glioma remains elusive, likely contributing to mixed outcomes in clinical trials. We delineated the phenotypic heterogeneity of TAMs across IDH-stratified gliomas. Notably, two TAM subsets with a mesenchymal phenotype were enriched in IDH-WT glioblastoma (GBM) and correlated with poorer patient survival and reduced response to anti-PD-1 immune checkpoint inhibitor (ICI). We proposed SLAMF9 receptor as a potential therapeutic target. Inference of gene regulatory networks identified PPARG, ELK1, and MXI1 as master transcription factors of mesenchymal BMD-TAMs. Our analyses of reciprocal tumor-TAM interactions revealed distinct crosstalk in IDH-WT tumors, including ANXA1-FPR1/3, FN1-ITGAVB1, VEGFA-NRP1, and TNFSF12-TNFRSF12A with known contribution to immunosuppression, tumor proliferation, invasion and TAM recruitment. Spatially resolved transcriptomics further elucidated the architectural organization of highlighted communications. Furthermore, we demonstrated significant upregulation of ANXA1, FN1, NRP1, and TNFRSF12A genes in IDH-WT tumors using bulk RNA-seq and RT-qPCR. Longitudinal expression analysis of candidate genes revealed no difference between primary and recurrent tumors indicating that the interactive network of malignant states with TAMs does not drastically change upon recurrence. Collectively, our study offers insights into the unique cellular composition and communication of TAMs in glioma TME, revealing novel vulnerabilities for therapeutic interventions in IDH-WT GBM.
Collapse
Affiliation(s)
- Meysam Motevasseli
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Darvishi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Khoshnevisan
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Zeinalizadeh
- Department of Neurosurgery, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Hiva Saffar
- Department of Pathology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Bayat
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Najafi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Abbaspour
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Mamivand
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Susan B Olson
- Molecular and Medical Genetics, Knight Diagnostics Laboratories, Oregon Health and Science University, Portland, OR, USA
| | - Mina Tabrizi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Molecular and Medical Genetics, Knight Diagnostics Laboratories, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
48
|
Oslund RC, Holland PM, Lesley SA, Fadeyi OO. Therapeutic potential of cis-targeting bispecific antibodies. Cell Chem Biol 2024; 31:1473-1489. [PMID: 39111317 DOI: 10.1016/j.chembiol.2024.07.004] [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: 12/14/2023] [Revised: 05/13/2024] [Accepted: 07/12/2024] [Indexed: 08/18/2024]
Abstract
The growing clinical success of bispecific antibodies (bsAbs) has led to rapid interest in leveraging dual targeting in order to generate novel modes of therapeutic action beyond mono-targeting approaches. While bsAbs that bind targets on two different cells (trans-targeting) are showing promise in the clinic, the co-targeting of two proteins on the same cell surface through cis-targeting bsAbs (cis-bsAbs) is an emerging strategy to elicit new functionalities. This includes the ability to induce proximity, enhance binding to a target, increase target/cell selectivity, and/or co-modulate function on the cell surface with the goal of altering, reversing, or eradicating abnormal cellular activity that contributes to disease. In this review, we focus on the impact of cis-bsAbs in the clinic, their emerging applications, and untangle the intricacies of improving bsAb discovery and development.
Collapse
|
49
|
AbuQeis I, Zou Y, Ba YC, Teeti AA. Neuroscience of cancer: Research progress and emerging of the field. IBRAIN 2024; 10:305-322. [PMID: 39346791 PMCID: PMC11427805 DOI: 10.1002/ibra.12172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 10/01/2024]
Abstract
Cancer cells immediately expand and penetrate adjoining tissues, as opposed to metastasis, that is the spread of cancer cells through the circulatory or lymphatic systems to more distant places via the invasion process. We found that a lack of studies discussed tumor development with the nervous system, by the aspects of cancer-tissue invasion (biological) and chemical modulation of growth that cascades by releasing neural-related factors from the nerve endings via chemical substances known as neurotransmitters. In this review, we aimed to carefully demonstrate and describe the cancer invasion and interaction with the nervous system, as well as reveal the research progress and the emerging neuroscience of cancer. An initial set of 160 references underwent systematic review and summarization. Through a meticulous screening process, these data were refined, ultimately leading to the inclusion of 98 studies that adhered to predetermined criteria. The outcomes show that one formidable challenge in the realm of cancer lies in its intrinsic heterogeneity and remarkable capacity for rapid adaptation. Despite advancements in genomics and precision medicine, there is still a need to identify new molecular targets. Considering cancer within its molecular and cellular environment, including neural components, is crucial for addressing this challenge. In conclusion, this review provides good referential data for direct, indirect, biological, and chemical interaction for nerve tissue-tumor interaction, suggesting the establishment of new therapy techniques and mechanisms by controlling and modifying neuron networks that supply signals to tumors.
Collapse
Affiliation(s)
- Issam AbuQeis
- Department of Radiology Palestinian Ministry of Health Ramallah Palestine
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Yu Zou
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Ying-Chun Ba
- Department of Anatomy, Institute of Neuroscience, School of Basic Medicine Kunming Medical University Kunming China
| | - Abeer A Teeti
- Department of Chemistry, School of Science Hebron University Hebron Palestine
- Department of Epidemiology, School of Public Health Kunming Medical University Kunming China
| |
Collapse
|
50
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [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: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|