101
|
Zhang N, Ye S, Wang X, Wang K, Zhong F, Yao F, Liu J, Huang B, Xu F, Wang X. Hepatic Symbiotic Bacterium L. reuteri FLRE5K1 Inhibits the Development and Progression of Hepatocellular Carcinoma via Activating the IFN-γ/CXCL10/CXCR3 Pathway. Probiotics Antimicrob Proteins 2024; 16:1158-1171. [PMID: 37289406 DOI: 10.1007/s12602-023-10098-y] [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] [Accepted: 05/25/2023] [Indexed: 06/09/2023]
Abstract
Symbiotic bacteria participate in the formation of the structure and function of the tissues and organs in which they live, and play an essential role in maintaining the balance between health and disease. Lactobacillus reuteri FLRE5K1 was isolated from the liver of healthy mice and proved to be a probiotic with anti-melanoma activity in previous studies. The relationship between hepatic symbiotic probiotics and hepatocellular carcinoma (HCC) has not been reported yet. In the present study, L. reuteri FLRE5K1 was initially confirmed to successfully enter the liver after being administered by gavage, and the efficacy of probiotic feeding on HCC and its potential mechanism of inhibiting tumor progression were investigated by an orthotopic liver cancer model established. The results showed that L. reuteri FLRE5K1 significantly reduced the tumor formation rate and inhibited tumor growth in mice. From the perspective of mechanism, activation of the IFN-γ/CXCL10/CXCR3 pathway, as well as its positive feedback on the secretion of IFN-γ, induced the polarization of Th0 cell to Th1 cells and inhibited the differentiation of Tregs, which played a key role in the inhibitory effect of L. reuteri FLRE5K1 on the development and progression of HCC.
Collapse
Affiliation(s)
- Nan Zhang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Shuiwen Ye
- Department of Blood Transfusion, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Xinlu Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Kang Wang
- Department of Traditional Chinese Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Fangyi Yao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Jing Liu
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China
| | - Feng Xu
- Jiangxi-Oai Joint Research Institute, Nanchang University, Nanchang, 330047, Jiangxi, China.
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Department of Clinical Laboratory, the Second Affiliated Hospital of Nanchang University, Nanchang, 330047, Jiangxi, China.
| |
Collapse
|
102
|
Reddy SU, Sadia FZ, Vancura A, Vancurova I. IFNγ-Induced Bcl3, PD-L1 and IL-8 Signaling in Ovarian Cancer: Mechanisms and Clinical Significance. Cancers (Basel) 2024; 16:2676. [PMID: 39123403 PMCID: PMC11311860 DOI: 10.3390/cancers16152676] [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: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
IFNγ, a pleiotropic cytokine produced not only by activated lymphocytes but also in response to cancer immunotherapies, has both antitumor and tumor-promoting functions. In ovarian cancer (OC) cells, the tumor-promoting functions of IFNγ are mediated by IFNγ-induced expression of Bcl3, PD-L1 and IL-8/CXCL8, which have long been known to have critical cellular functions as a proto-oncogene, an immune checkpoint ligand and a chemoattractant, respectively. However, overwhelming evidence has demonstrated that these three genes have tumor-promoting roles far beyond their originally identified functions. These tumor-promoting mechanisms include increased cancer cell proliferation, invasion, angiogenesis, metastasis, resistance to chemotherapy and immune escape. Recent studies have shown that IFNγ-induced Bcl3, PD-L1 and IL-8 expression is regulated by the same JAK1/STAT1 signaling pathway: IFNγ induces the expression of Bcl3, which then promotes the expression of PD-L1 and IL-8 in OC cells, resulting in their increased proliferation and migration. In this review, we summarize the recent findings on how IFNγ affects the tumor microenvironment and promotes tumor progression, with a special focus on ovarian cancer and on Bcl3, PD-L1 and IL-8/CXCL8 signaling. We also discuss promising novel combinatorial strategies in clinical trials targeting Bcl3, PD-L1 and IL-8 to increase the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Ivana Vancurova
- Department of Biological Sciences, St. John’s University, New York, NY 11439, USA; (S.U.R.); (F.Z.S.); (A.V.)
| |
Collapse
|
103
|
Park H, Song J, Jeong HW, Grönloh MLB, Koh BI, Bovay E, Kim KP, Klotz L, Thistlethwaite PA, van Buul JD, Sorokin L, Adams RH. Apelin modulates inflammation and leukocyte recruitment in experimental autoimmune encephalomyelitis. Nat Commun 2024; 15:6282. [PMID: 39060233 PMCID: PMC11282314 DOI: 10.1038/s41467-024-50540-5] [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: 07/21/2023] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Demyelination due to autoreactive T cells and inflammation in the central nervous system are principal features of multiple sclerosis (MS), a chronic and highly disabling human disease affecting brain and spinal cord. Here, we show that treatment with apelin, a secreted peptide ligand for the G protein-coupled receptor APJ/Aplnr, is protective in experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Apelin reduces immune cell entry into the brain, delays the onset and reduces the severity of EAE. Apelin affects the trafficking of leukocytes through the lung by modulating the expression of cell adhesion molecules that mediate leukocyte recruitment. In addition, apelin induces the internalization and desensitization of its receptor in endothelial cells (ECs). Accordingly, protection against EAE major outcomes of apelin treatment are phenocopied by loss of APJ/Aplnr function, achieved by EC-specific gene inactivation in mice or knockdown experiments in cultured primary endothelial cells. Our findings highlight the importance of the lung-brain axis in neuroinflammation and indicate that apelin targets the transendothelial migration of immune cells into the lung during acute inflammation.
Collapse
Affiliation(s)
- Hongryeol Park
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany.
| | - Jian Song
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Max L B Grönloh
- Vascular Cell Biology Lab, Department of Medical Biochemistry, Amsterdam UMC, and Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Bong Ihn Koh
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Esther Bovay
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany
| | - Kee-Pyo Kim
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Luisa Klotz
- Department of Neurology, University of Münster, Münster, Germany
| | | | - Jaap D van Buul
- Vascular Cell Biology Lab, Department of Medical Biochemistry, Amsterdam UMC, and Section Molecular Cytology at Swammerdam Institute for Life Sciences, Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry and Cells-in-Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, Münster, Germany.
| |
Collapse
|
104
|
Tokito T, Kolesnik O, Sørensen J, Artac M, Quintela ML, Lee JS, Hussein M, Pless M, Paz-Ares L, Leopold L, Daniel J, Munteanu M, Samkari A, Xu L, Butts C. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab as first-line treatment for metastatic non-small cell lung cancer with high levels of programmed death-ligand 1: a randomized, double-blind phase 2 study. BMC Cancer 2024; 23:1251. [PMID: 39054476 PMCID: PMC11270761 DOI: 10.1186/s12885-023-11203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 07/19/2023] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Pembrolizumab is a first-line therapy for certain patients with advanced/metastatic non-small cell lung cancer (NSCLC). Combining pembrolizumab with other immunotherapies may enhance tumor cell killing and clinical outcomes. Epacadostat is a selective inhibitor of indoleamine 2,3-dioxygenase 1, an immuno-regulatory enzyme involved in tryptophan to kynurenine metabolism that inhibits T cell-mediated immune responses. METHODS In this randomized phase II study, patients with metastatic NSCLC expressing high (≥ 50%) programmed death-ligand 1 (PD-L1) levels received pembrolizumab 200 mg every 21 days plus oral epacadostat 100 mg twice daily (combination) or matching placebo (control). The primary objective was objective response rate (ORR); secondary objectives were progression-free survival (PFS), overall survival (OS), duration of response (DOR) and safety/tolerability. RESULTS 154 patients were randomized (77 per group). Median (range) follow-up was 6.8 months (0.1-11.4) and 7.0 months (0.2-11.9) in the combination and control groups, respectively Confirmed ORR was similar between groups (combination: 32.5%, 95% CI 22.2-44.1; control: 39.0%, 95% CI 28.0-50.8; difference: - 6.5, 95% CI - 21.5 to 8.7; 1-sided P = 0.8000). Median (range) DOR was 6.2 months (1.9 + to 6.5 +) and not reached (1.9 + to 8.6 +) in the combination and control groups, respectively. Although not formally tested, median PFS was 6.7 and 6.2 months for the combination and control groups, respectively, and median OS was not reached in either group. Circulating kynurenine levels increased from C1D1 to C2D1 (P < 0.01) in the control group and decreased from C1D1 to C2D1 (P < 0.01) in the combination group but were not normalized in most patients. The most frequent serious adverse events (AEs) (≥ 2%) were pneumonia (4.0%), anemia (2.7%), atelectasis (2.7%) and pneumonitis (2.7%) in the combination group and pneumonia (3.9%), pneumonitis (2.6%) and hypotension (2.6%) in the control group. Two deaths due to drug-related AEs were reported, both in the control group. CONCLUSIONS Addition of epacadostat to pembrolizumab therapy for PD-L1-high metastatic NSCLC was generally well tolerated but did not demonstrate an improved therapeutic effect. Evaluating higher doses of epacadostat that normalize kynurenine levels when given in combination with checkpoint inhibitors may be warranted. TRIAL REGISTRATION ClinicalTrials.gov, NCT03322540. Registered 10/26/2017.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Female
- Humans
- Male
- Middle Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- B7-H1 Antigen/antagonists & inhibitors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Double-Blind Method
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Oximes/administration & dosage
- Oximes/therapeutic use
- Oximes/adverse effects
- Progression-Free Survival
- Sulfonamides/administration & dosage
- Sulfonamides/therapeutic use
- Sulfonamides/adverse effects
- Neoplasm Metastasis
- Placebos
Collapse
Affiliation(s)
- Takaaki Tokito
- Division of Respirology, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan.
| | - Oleksii Kolesnik
- Zaporizhzhya Regional Clinical Oncology Center, Zaporizhzhya State Medical University, Zaporozhye, Ukraine
| | - Jens Sørensen
- Department of Oncology, Centre for Cancer and Organ Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Mehmet Artac
- Department of Medical Oncology, Necmettin Erbakan Universitesi Meram Tip Fakultesi Hastanesi, Konya, Turkey
| | - Martín Lázaro Quintela
- Department of Medical Oncology, Hospital Álvaro Cunqueiro-Complexo Hospitalario Universitario de Vigo, Vigo, Spain
| | - Jong-Seok Lee
- Division of Hematology-Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | | | - Miklos Pless
- Department of Medical Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario, 12 de Octubre, Universidad Complutense & Ciberonc, Madrid, Spain
| | | | | | | | | | - Lu Xu
- Merck & Co., Inc., Rahway, NJ, United States
| | - Charles Butts
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, Edmonton, Canada
| |
Collapse
|
105
|
Lu KYF, Alqaderi H, Bin Hasan S, Alhazmi H, Alghounaim M, Devarajan S, Freire M, Altabtbaei K. Sputum production and salivary microbiome in COVID-19 patients reveals oral-lung axis. PLoS One 2024; 19:e0300408. [PMID: 39052548 PMCID: PMC11271936 DOI: 10.1371/journal.pone.0300408] [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: 03/18/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
SARS-CoV-2, a severe respiratory disease primarily targeting the lungs, was the leading cause of death worldwide during the pandemic. Understanding the interplay between the oral microbiome and inflammatory cytokines during acute infection is crucial for elucidating host immune responses. This study aimed to explore the relationship between the oral microbiome and cytokines in COVID-19 patients, particularly those with and without sputum production. Saliva and blood samples from 50 COVID-19 patients were subjected to 16S ribosomal RNA gene sequencing for oral microbiome analysis, and 65 saliva and serum cytokines were assessed using Luminex multiplex analysis. The Mann-Whitney test was used to compare cytokine levels between individuals with and without sputum production. Logistic regression machine learning models were employed to evaluate the predictive capability of oral microbiome, salivary, and blood biomarkers for sputum production. Significant differences were observed in the membership (Jaccard dissimilarity: p = 0.016) and abundance (PhILR dissimilarity: p = 0.048; metagenomeSeq) of salivary microbial communities between patients with and without sputum production. Seven bacterial genera, including Prevotella, Streptococcus, Actinomyces, Atopobium, Filifactor, Leptotrichia, and Selenomonas, were more prevalent in patients with sputum production (p<0.05, Fisher's exact test). Nine genera, including Prevotella, Megasphaera, Stomatobaculum, Selenomonas, Leptotrichia, Veillonella, Actinomyces, Atopobium, and Corynebacteria, were significantly more abundant in the sputum-producing group, while Lachnoanaerobaculum was more prevalent in the non-sputum-producing group (p<0.05, ANCOM-BC). Positive correlations were found between salivary IFN-gamma and Eotaxin2/CCL24 with sputum production, while negative correlations were noted with serum MCP3/CCL7, MIG/CXCL9, IL1 beta, and SCF (p<0.05, Mann-Whitney test). The machine learning model using only oral bacteria input outperformed the model that included all data: blood and saliva biomarkers, as well as clinical and demographic variables, in predicting sputum production in COVID-19 subjects. The performance metrics were as follows, comparing the model with only bacteria input versus the model with all input variables: precision (95% vs. 75%), recall (100% vs. 50%), F1-score (98% vs. 60%), and accuracy (82% vs. 66%).
Collapse
Affiliation(s)
- Korina Yun-Fan Lu
- Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Hend Alqaderi
- Tufts University School of Dental Medicine, Boston, Massachusetts, United States of America
- Dasman Diabetes Institute, Dasman, Kuwait
| | | | - Hesham Alhazmi
- Harvard School of Dental Medicine, Boston, Massachusetts, United States of America
- Department of Preventive Dentistry, Division of Pediatric Dentistry, Umm Al-Qura University, Mekkah, Saudi Arabia
| | | | | | - Marcelo Freire
- Department of Genomic Medicine and Infectious Diseases, J. Craig Venter Institute, La Jolla, California, United States of America
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, United States of America
| | | |
Collapse
|
106
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
107
|
Lu MJ, Zhang JQ, Nie ZY, Yan TH, Cao YB, Zhang LC, Li L. Monocyte/macrophage-mediated venous thrombus resolution. Front Immunol 2024; 15:1429523. [PMID: 39100675 PMCID: PMC11297357 DOI: 10.3389/fimmu.2024.1429523] [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: 05/08/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Venous thromboembolism (VTE) poses a notable risk of morbidity and mortality. The natural resolution of the venous thrombus might be a potential alternative treatment strategy for VTE. Monocytes/macrophages merge as pivotal cell types in the gradual resolution of the thrombus. In this review, the vital role of macrophages in inducing inflammatory response, augmenting neovascularization, and facilitating the degradation of fibrin and collagen during thrombus resolution was described. The two phenotypes of macrophages involved in thrombus resolution and their dual functions were discussed. Macrophages expressing various factors, including cytokines and their receptors, adhesion molecules, chemokine receptors, vascular endothelial growth factor receptors, profibrinolytic- or antifibrinolytic-related enzymes, and other elements, are explored for their potential to promote or attenuate thrombus resolution. Furthermore, this review provides a comprehensive summary of new and promising therapeutic candidate drugs associated with monocytes/macrophages that have been demonstrated to promote or impair thrombus resolution. However, further clinical trials are essential to validate their efficacy in VTE therapy.
Collapse
Affiliation(s)
- Meng-Jiao Lu
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Jia-Qi Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhou-Yu Nie
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Hua Yan
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Yong-Bing Cao
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM- Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
108
|
Pesini C, Artal L, Paúl Bernal J, Sánchez Martinez D, Pardo J, Ramírez-Labrada A. In-depth analysis of the interplay between oncogenic mutations and NK cell-mediated cancer surveillance in solid tumors. Oncoimmunology 2024; 13:2379062. [PMID: 39036370 PMCID: PMC11259085 DOI: 10.1080/2162402x.2024.2379062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Natural killer (NK) cells play a crucial role in antitumoral and antiviral responses. Yet, cancer cells can alter themselves or the microenvironment through the secretion of cytokines or other factors, hindering NK cell activation and promoting a less cytotoxic phenotype. These resistance mechanisms, often referred to as the "hallmarks of cancer" are significantly influenced by the activation of oncogenes, impacting most, if not all, of the described hallmarks. Along with oncogenes, other types of genes, the tumor suppressor genes are frequently mutated or modified during cancer. Traditionally, these genes have been associated with uncontrollable tumor growth and apoptosis resistance. Recent evidence suggests oncogenic mutations extend beyond modulating cell death/proliferation programs, influencing cancer immunosurveillance. While T cells have been more studied, the results obtained highlight NK cells as emerging key protagonists for enhancing tumor cell elimination by modulating oncogenic activity. A few recent studies highlight the crucial role of oncogenic mutations in NK cell-mediated cancer recognition, impacting angiogenesis, stress ligands, and signaling balance within the tumor microenvironment. This review will critically examine recent discoveries correlating oncogenic mutations to NK cell-mediated cancer immunosurveillance, a relatively underexplored area, particularly in the era dominated by immune checkpoint inhibitors and CAR-T cells. Building on these insights, we will explore opportunities to improve NK cell-based immunotherapies, which are increasingly recognized as promising alternatives for treating low-antigenic tumors, offering significant advantages in terms of safety and manufacturing suitability.
Collapse
Affiliation(s)
- Cecilia Pesini
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Laura Artal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Institute of Carbochemistry (ICB-CSIC), Zaragoza, Spain
| | - Jorge Paúl Bernal
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Diego Sánchez Martinez
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Aragón I + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain
| | - Julián Pardo
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
- Department of Microbiology, Radiology, Pediatry and Public Health, University of Zaragoza, Zaragoza, Spain
| | - Ariel Ramírez-Labrada
- Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
- Center for Biomedical Research in the Network of Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Zaragoza, Spain
| |
Collapse
|
109
|
Maury AA, Holton KF. Biomarkers Associated with Depression Improvement in Veterans with Gulf War Illness Using the Low-Glutamate Diet. Nutrients 2024; 16:2255. [PMID: 39064698 PMCID: PMC11280460 DOI: 10.3390/nu16142255] [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: 06/11/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Gulf War Illness (GWI) is a chronic multi-symptom neurological disorder affecting veterans of the Gulf War that is commonly comorbid with depression. A secondary data analysis was conducted to examine serum homocysteine and inflammatory cytokines (IFN-γ, IL-6, IL-1β, TNF-α) as potential biomarkers of depression improvement among veterans with GWI after a one-month dietary intervention aimed at reducing excitotoxicity and increasing micronutrients. Analyses, including multiple linear and logistic regression, were conducted in R studio. Dietary adherence was estimated using a specialized excitotoxin food frequency questionnaire (FFQ), and depression was measured using the Center for Epidemiologic Studies Depression (CES-D) scale. After one month on the diet, 52% of participants reported a significant decrease in depression (p < 0.01). Greater dietary adherence (FFQ) was associated with increased likelihood of depression improvement; OR (95% CI) = 1.06 (1.01, 1.11), (p = 0.02). Reduced homocysteine was associated with depression improvement after adjusting for FFQ change (β = 2.58, p = 0.04), and serum folate and vitamin B12 were not mediators of this association. Reduction in IFN-γ was marginally associated with likelihood of depression improvement (OR (95% CI) = 1.11 (0.00, 1.42), (p = 0.06)), after adjustment for potential confounders. Findings suggest that homocysteine, and possibly IFN-γ, may serve as biomarkers for depression improvement in GWI. Larger trials are needed to replicate this work.
Collapse
Affiliation(s)
- Amy A. Maury
- Department of Neuroscience, American University, Washington, DC 20016, USA;
| | - Kathleen F. Holton
- Department of Health Studies, American University, Washington, DC 20016, USA
- Center for Neuroscience and Behavior, American University, Washington, DC 20016, USA
| |
Collapse
|
110
|
Sinmez CC, Tüfekçi E, Demir BŞ, Eken A, Guneş V, Ekici S, Bozkaya E, Aykun Aİ. Investigation of immunomodulatory and cytotoxic effects of shed snake skin ( Elaphe sauromates) extract. Front Pharmacol 2024; 15:1270970. [PMID: 39070782 PMCID: PMC11272602 DOI: 10.3389/fphar.2024.1270970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 06/11/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction Shed snake skin (SSS) is commonly used empirically in ethnomedicine to treat psoriasis, acne, warts, eczema, scabies, open wounds, hemorrhoids, and glaucoma. Although a few studies exist, SSS extracts' in vitro immunological effects have yet to be well described. Therefore, we aimed to investigate the immunomodulatory effects of SSS extract on murine lymphocytes and T cells. Methods Hexane, methanol, and chloroform extractions were conducted in collected SSS samples. Protein concentrations in the SSS extract were measured. The cytotoxic and anticancer activities were measured using L929 Fibroblast and SK MEL 30 Cell Lines via MTT assay as described in TS EN ISO 10993-5. Immunomodulatory activities of SSS extract on total lymphocytes or enriched CD4+ T cell cultures, their cell-specific pro-inflammatory cytokines (IL-6, IL-1β. IL-12p40, IL-23p19, TNF-α, IL-17A, IFN-γ, IL-10, TGFβ1) levels were measured via FACS ARIA III analysis and related gene expression with Real-Time Quantitative Polymerase Chain Reaction (Rt-qPCR). Results Hexane, methanol, and chloroform-extracted SSS were tested on SK-MEL-30 cells via MTT and revealed a superior anti-proliferative effect for hexane extract of SSS at low concentrations. SSS treatment of murine lymphocytes augmented Tnf-α and IFN-γ levels produced by CD3+ T cells when lymphocytes were activated with anti-CD3/CD28 or LPS stimulation. This effect required the presence of non-T cells, possibly antigen-presenting cells, and was not observed on purified CD4+ T cells. Additionally, SSS significantly blocked suppressive cytokine Tgfb gene expression (but not Il10) without altering in vitro Treg generation/or expansion. Discussion This is the first in vitro study investigating SSS's anti-tumor and immunomodulatory effects. Our data provide evidence for SSS's anti-proliferative activity on SK-MEL-30 cells and its pro-inflammatory role on murine lymphocytes, which warrants further investigation of the potential use of SSS extract with in vivo disease models.
Collapse
Affiliation(s)
- Cagri Caglar Sinmez
- Department of History of Veterinary Medicine and Deontology, Faculty of Veterinary Medicine, Ziya Eren Drug Research and Application Center (ERFARMA), Erciyes University, Kayseri, Türkiye
| | - Emre Tüfekçi
- Department of Internal Medicine, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Türkiye
| | - Büşra Şeniz Demir
- Department of Medical Biology, School of Medicine, Betül-Ziya Eren Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Türkiye
| | - Ahmet Eken
- Department of Medical Biology, School of Medicine, Betül-Ziya Eren Genome and Stem Cell Center (GENKÖK), Erciyes University, Kayseri, Türkiye
| | - Vehbi Guneş
- Department of Internal Medicine, Faculty of Veterinary Medicine, Experimental Research and Application Center (DEKAM), Kayseri, Türkiye
| | - Seda Ekici
- Veterinary Control Central Research Institute, Ankara, Türkiye
| | - Esra Bozkaya
- Scientific and Technological Research Application and Research Center, Kırıkkale University, Kırıkkale, Türkiye
| | - Ali İlteriş Aykun
- Department of History of Veterinary Medicine and Deontology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Türkiye
| |
Collapse
|
111
|
Ng BD, Rajagopalan A, Kousa AI, Fischman JS, Chen S, Massa A, Elias HK, Manuele D, Galiano M, Lemarquis AL, Boardman AP, DeWolf S, Pierce J, Bogen B, James SE, van den Brink MRM. IL-18-secreting multiantigen targeting CAR T cells eliminate antigen-low myeloma in an immunocompetent mouse model. Blood 2024; 144:171-186. [PMID: 38579288 PMCID: PMC11302468 DOI: 10.1182/blood.2023022293] [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: 09/15/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Multiple myeloma is a plasma cell malignancy that is currently incurable with conventional therapies. Following the success of CD19-targeted chimeric antigen receptor (CAR) T cells in leukemia and lymphoma, CAR T cells targeting B-cell maturation antigen (BCMA) more recently demonstrated impressive activity in relapsed and refractory myeloma patients. However, BCMA-directed therapy can fail due to weak expression of BCMA on myeloma cells, suggesting that novel approaches to better address this antigen-low disease may improve patient outcomes. We hypothesized that engineered secretion of the proinflammatory cytokine interleukin-18 (IL-18) and multiantigen targeting could improve CAR T-cell activity against BCMA-low myeloma. In a syngeneic murine model of myeloma, CAR T cells targeting the myeloma-associated antigens BCMA and B-cell activating factor receptor (BAFF-R) failed to eliminate myeloma when these antigens were weakly expressed, whereas IL-18-secreting CAR T cells targeting these antigens promoted myeloma clearance. IL-18-secreting CAR T cells developed an effector-like T-cell phenotype, promoted interferon-gamma production, reprogrammed the myeloma bone marrow microenvironment through type-I/II interferon signaling, and activated macrophages to mediate antimyeloma activity. Simultaneous targeting of weakly-expressed BCMA and BAFF-R with dual-CAR T cells enhanced T-cell:target-cell avidity, increased overall CAR signal strength, and stimulated antimyeloma activity. Dual-antigen targeting augmented CAR T-cell secretion of engineered IL-18 and facilitated elimination of larger myeloma burdens in vivo. Our results demonstrate that combination of engineered IL-18 secretion and multiantigen targeting can eliminate myeloma with weak antigen expression through distinct mechanisms.
Collapse
Affiliation(s)
- Brandon D. Ng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pharmacology, Weill Cornell Medicine, New York, NY
| | - Adhithi Rajagopalan
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anastasia I. Kousa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Jacob S. Fischman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA
| | - Sophia Chen
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alyssa Massa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Harold K. Elias
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dylan Manuele
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Michael Galiano
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andri L. Lemarquis
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander P. Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan DeWolf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonah Pierce
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
| | | | - Scott E. James
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R. M. van den Brink
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
112
|
Teng HW, Wang TY, Lin CC, Tong ZJ, Cheng HW, Wang HT. Interferon Gamma Induces Higher Neutrophil Extracellular Traps Leading to Tumor-Killing Activity in Microsatellite Stable Colorectal Cancer. Mol Cancer Ther 2024; 23:1043-1056. [PMID: 38346939 DOI: 10.1158/1535-7163.mct-23-0744] [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: 11/14/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 07/03/2024]
Abstract
Many patients with colorectal cancer do not respond to immune checkpoint blockade (ICB) therapy, highlighting the urgent need to understand tumor resistance mechanisms. Recently, the link between the IFNγ signaling pathway integrity and ICB resistance in the colorectal cancer tumor microenvironment has been revealed. The immunosuppressive microenvironment poses a significant challenge to antitumor immunity in colorectal cancer development. Tumor-associated neutrophils found in tumor tissues exhibit an immunosuppressive phenotype and are associated with colorectal cancer patient prognosis. Neutrophil extracellular traps (NET), DNA meshes containing cytotoxic enzymes released into the extracellular space, may be promising therapeutic targets in cancer. This study showed increased NETs in tumor tissues and peripheral neutrophils of high levels of microsatellite instability (MSI-H) patients with colorectal cancer compared with microsatellite stable (MSS) patients with colorectal cancer. IFNγ response genes were enriched in MSI-H patients with colorectal cancer compared with patients with MSS colorectal cancer. Co-culturing neutrophils with MSI-H colorectal cancer cell lines induced more NET formation and higher cellular apoptosis than MSS colorectal cancer cell lines. IFNγ treatment induced more NET formation and apoptosis in MSS colorectal cancer cell lines. Using subcutaneous or orthotopic CT-26 (MSS) tumor-bearing mice models, IFNγ reduced tumor size and enhanced PD-1 antibody-induced tumor-killing activity, accompanied by upregulated NETs and cellular apoptosis. These findings suggest that IFNγ could be a therapeutic strategy for MSS colorectal cancer.
Collapse
Affiliation(s)
- Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tean-Ya Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chi Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctor degree program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
113
|
de Winter N, Ji J, Sintou A, Forte E, Lee M, Noseda M, Li A, Koenig AL, Lavine KJ, Hayat S, Rosenthal N, Emanueli C, Srivastava PK, Sattler S. Persistent transcriptional changes in cardiac adaptive immune cells following myocardial infarction: New evidence from the re-analysis of publicly available single cell and nuclei RNA-sequencing data sets. J Mol Cell Cardiol 2024; 192:48-64. [PMID: 38734060 DOI: 10.1016/j.yjmcc.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
INTRODUCTION Chronic immunopathology contributes to the development of heart failure after a myocardial infarction. Both T and B cells of the adaptive immune system are present in the myocardium and have been suggested to be involved in post-MI immunopathology. METHODS We analyzed the B and T cell populations isolated from previously published single cell RNA-sequencing data sets (PMID: 32130914, PMID: 35948637, PMID: 32971526 and PMID: 35926050), of the mouse and human heart, using differential expression analysis, functional enrichment analysis, gene regulatory inferences, and integration with autoimmune and cardiovascular GWAS. RESULTS Already at baseline, mature effector B and T cells are present in the human and mouse heart, having increased activity in transcription factors maintaining tolerance (e.g. DEAF1, JDP2, SPI-B). Following MI, T cells upregulate pro-inflammatory transcript levels (e.g. Cd11, Gzmk, Prf1), while B cells upregulate activation markers (e.g. Il6, Il1rn, Ccl6) and collagen (e.g. Col5a2, Col4a1, Col1a2). Importantly, pro-inflammatory and fibrotic transcription factors (e.g. NFKB1, CREM, REL) remain active in T cells, while B cells maintain elevated activity in transcription factors related to immunoglobulin production (e.g. ERG, REL) in both mouse and human post-MI hearts. Notably, genes differentially expressed in post-MI T and B cells are associated with cardiovascular and autoimmune disease. CONCLUSION These findings highlight the varied and time-dependent dynamic roles of post-MI T and B cells. They appear ready-to-go and are activated immediately after MI, thus participate in the acute wound healing response. However, they subsequently remain in a state of pro-inflammatory activation contributing to persistent immunopathology.
Collapse
Affiliation(s)
- Natasha de Winter
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Jiahui Ji
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Amalia Sintou
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, United States
| | - Michael Lee
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Michela Noseda
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; British Heart Foundation Centre For Research Excellence, Imperial College London, United Kingdom
| | - Aoxue Li
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andrew L Koenig
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States
| | - Kory J Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States
| | | | - Nadia Rosenthal
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; The Jackson Laboratory, Bar Harbor, United States
| | - Costanza Emanueli
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; British Heart Foundation Centre For Research Excellence, Imperial College London, United Kingdom
| | - Prashant K Srivastava
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Susanne Sattler
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; Department of Cardiology, Medical University of Graz, Austria; Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria.
| |
Collapse
|
114
|
Samman N, Mohabatkar H, Behbahani M, Ganjlikhani Hakemi M. Bioinformatics design of a peptide vaccine containing sarcoma antigen NY-SAR-35 epitopes against breast cancer and evaluation of its immunological function in BALB/c mouse model. PLoS One 2024; 19:e0306117. [PMID: 38923980 PMCID: PMC11207152 DOI: 10.1371/journal.pone.0306117] [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: 01/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
The development of a cancer vaccine has become an essential focus in the field of medical biotechnology and immunology. In our study, the NY-SAR-35 cancer/testis antigen was targeted to design a novel peptide vaccine using bioinformatics tools, and BALB/c mice were used to evaluate the vaccine's immunological function. This evaluation involved assessing peptide-specific IgG levels in the serum via ELISA and measuring the levels of IFN-γ, IL-4, and granzyme B in the supernatant of cultured splenocytes. The final vaccine construct consisted of two T lymphocyte epitopes linked by the AAY linker. This construct displayed high antigenicity, non-allergenicity, non-toxicity, stability, and ability to induce IFN-γ and IL-4. It showed stable dynamics with both human MHC-I and II molecules, as well as mouse MHC-II molecules, and revealed strong Van der Waals and electrostatic energies. Emulsifying our peptide vaccine in incomplete Freund's adjuvant resulted in a remarkable increase in the levels of IgG. The splenocytes of mice that received the combination of peptide and adjuvant displayed a noteworthy increase in IFN-γ, IL-4, and granzyme B secretion. Additionally, their lymphocytes exhibited higher proliferation rates compared to the control group. Our data demonstrated that our vaccine could stimulate a robust immune response, making it a promising candidate for cancer prevention. However, clinical trials are necessary to assess its efficacy in humans.
Collapse
Affiliation(s)
- Nour Samman
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Hassan Mohabatkar
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mandana Behbahani
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mazdak Ganjlikhani Hakemi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
115
|
Purbey PK, Seo J, Paul MK, Iwamoto KS, Daly AE, Feng AC, Champhekar AS, Langerman J, Campbell KM, Schaue D, McBride WH, Dubinett SM, Ribas A, Smale ST, Scumpia PO. Opposing tumor-cell-intrinsic and -extrinsic roles of the IRF1 transcription factor in antitumor immunity. Cell Rep 2024; 43:114289. [PMID: 38833371 PMCID: PMC11315447 DOI: 10.1016/j.celrep.2024.114289] [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: 11/30/2023] [Revised: 03/13/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024] Open
Abstract
Type I interferon (IFN-I) and IFN-γ foster antitumor immunity by facilitating T cell responses. Paradoxically, IFNs may promote T cell exhaustion by activating immune checkpoints. The downstream regulators of these disparate responses are incompletely understood. Here, we describe how interferon regulatory factor 1 (IRF1) orchestrates these opposing effects of IFNs. IRF1 expression in tumor cells blocks Toll-like receptor- and IFN-I-dependent host antitumor immunity by preventing interferon-stimulated gene (ISG) and effector programs in immune cells. In contrast, expression of IRF1 in the host is required for antitumor immunity. Mechanistically, IRF1 binds distinctly or together with STAT1 at promoters of immunosuppressive but not immunostimulatory ISGs in tumor cells. Overexpression of programmed cell death ligand 1 (PD-L1) in Irf1-/- tumors only partially restores tumor growth, suggesting multifactorial effects of IRF1 on antitumor immunity. Thus, we identify that IRF1 expression in tumor cells opposes host IFN-I- and IRF1-dependent antitumor immunity to facilitate immune escape and tumor growth.
Collapse
Affiliation(s)
- Prabhat K Purbey
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - Joowon Seo
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Manash K Paul
- Department of Medicine, Division of Pulmonology and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Keisuke S Iwamoto
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Allison E Daly
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - An-Chieh Feng
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ameya S Champhekar
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Justin Langerman
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Katie M Campbell
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - William H McBride
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Steven M Dubinett
- Department of Medicine, Division of Pulmonology and Critical Care Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Stephen T Smale
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Howard Hughes Medical Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Philip O Scumpia
- Department of Medicine, Division of Dermatology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA; VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| |
Collapse
|
116
|
Hasan A, Alonazi WB, Ibrahim M, Bin L. Immunoinformatics and Reverse Vaccinology Approach for the Identification of Potential Vaccine Candidates against Vandammella animalimors. Microorganisms 2024; 12:1270. [PMID: 39065039 PMCID: PMC11278545 DOI: 10.3390/microorganisms12071270] [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: 04/29/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Vandammella animalimorsus is a Gram-negative and non-motile bacterium typically transmitted to humans through direct contact with the saliva of infected animals, primarily through biting, scratches, or licks on fractured skin. The absence of a confirmed post-exposure treatment of V. animalimorsus bacterium highlights the imperative for developing an effective vaccine. We intended to determine potential vaccine candidates and paradigm a chimeric vaccine against V. animalimorsus by accessible public data analysis of the strain by utilizing reverse vaccinology. By subtractive genomics, five outer membranes were prioritized as potential vaccine candidates out of 2590 proteins. Based on the instability index and transmembrane helices, a multidrug transporter protein with locus ID A0A2A2AHJ4 was designated as a potential candidate for vaccine construct. Sixteen immunodominant epitopes were retrieved by utilizing the Immune Epitope Database. The epitope encodes the strong binding affinity, nonallergenic properties, non-toxicity, high antigenicity scores, and high solubility revealing the more appropriate vaccine construct. By utilizing appropriate linkers and adjuvants alongside a suitable adjuvant molecule, the epitopes were integrated into a chimeric vaccine to enhance immunogenicity, successfully eliciting both adaptive and innate immune responses. Moreover, the promising physicochemical features, the binding confirmation of the vaccine to the major innate immune receptor TLR-4, and molecular dynamics simulations of the designed vaccine have revealed the promising potential of the selected candidate. The integration of computational methods and omics data has demonstrated significant advantages in discovering novel vaccine targets and mitigating vaccine failure rates during clinical trials in recent years.
Collapse
Affiliation(s)
- Ahmad Hasan
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| | - Wadi B. Alonazi
- Health Administration Department, College of Business Administration, King Saud University, Riyadh 11421, Saudi Arabia;
| | - Muhammad Ibrahim
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| | - Li Bin
- State Key Laboratory of Rice Biology and Breeding, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Institute of Biotechnology, Zhejiang University, Hangzhou 310058, China; (A.H.); (M.I.)
| |
Collapse
|
117
|
Heim C, Hartig L, Weinelt N, Moser LM, Salzmann-Manrique E, Merker M, Wels WS, Tonn T, Bader P, Klusmann JH, van Wijk SJ, Rettinger E. Bortezomib promotes the TRAIL-mediated killing of resistant rhabdomyosarcoma by ErbB2/Her2-targeted CAR-NK-92 cells via DR5 upregulation. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200802. [PMID: 38706988 PMCID: PMC11067460 DOI: 10.1016/j.omton.2024.200802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
Treatment resistance and immune escape are hallmarks of metastatic rhabdomyosarcoma (RMS), underscoring the urgent medical need for therapeutic agents against this disease entity as a key challenge in pediatric oncology. Chimeric antigen receptor (CAR)-based immunotherapies, such as the ErbB2 (Her2)-CAR-engineered natural killer (NK) cell line NK-92/5.28.z, provide antitumor cytotoxicity primarily through CAR-mediated cytotoxic granule release and thereafter-even in cases with low surface antigen expression or tumor escape-by triggering intrinsic NK cell-mediated apoptosis induction via additional ligand/receptors. In this study, we showed that bortezomib increased susceptibility toward apoptosis in clinically relevant RMS cell lines RH30 and RH41, and patient-derived RMS tumor organoid RMS335, by upregulation of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor DR5 in these metastatic, relapsed/refractory (r/r) RMS tumors. Subsequent administration of NK-92/5.28.z cells significantly enhanced antitumor activity in vitro. Applying recombinant TRAIL instead of NK-92/5.28.z cells confirmed that the synergistic antitumor effects of the combination treatment were mediated via TRAIL. Western blot analyses indicated that the combination treatment with bortezomib and NK-92/5.28.z cells increased apoptosis by interacting with the nuclear factor κB, JNK, and caspase pathways. Overall, bortezomib pretreatment can sensitize r/r RMS tumors to CAR- and, by upregulating DR5, TRAIL-mediated cytotoxicity of NK-92/5.28.z cells.
Collapse
Affiliation(s)
- Catrin Heim
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Leonie Hartig
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Nadine Weinelt
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
| | - Laura M. Moser
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Michael Merker
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Torsten Tonn
- DRK-Blutspendedienst Baden-Württemberg/Hessen gemeinnützige GmbH, 60505 Frankfurt am Main, Germany
| | - Peter Bader
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Jan-Henning Klusmann
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Sjoerd J.L. van Wijk
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| | - Eva Rettinger
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| |
Collapse
|
118
|
Luo D, Gong Z, Zhan Q, Lin S. Causal association of circulating cytokines with the risk of lung cancer: a Mendelian randomization study. Front Oncol 2024; 14:1373380. [PMID: 38957317 PMCID: PMC11217496 DOI: 10.3389/fonc.2024.1373380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
Background Lung cancer is the deadliest and most prevalent malignancy worldwide. While smoking is an established cause, evidence to identify other causal factors remains lacking. Current research indicates chronic inflammation is involved in tumorigenesis and cancer development, though the specific mechanisms underlying the role of inflammatory cytokines in lung cancer pathogenesis remain unclear. This study implemented Mendelian randomization (MR) analysis to investigate the causal effects of circulating cytokines on lung cancer development. Methods We performed a two-sample MR analysis in Europeans utilizing publicly available genome-wide association study summary statistics. Single nucleotide polymorphisms significantly associated with cytokine were selected as genetic instrumental variables. Results Genetically predicted levels of the chemokine interleukin-18 (IL-18) (OR = 0.942, 95% CI: 0.897-0.990, P = 0.018) exerted significant negative causal effects on overall lung cancer risk in this analysis. Examining specific histologic subtypes revealed further evidence of genetic associations. Stem cell factor (SCF) (OR = 1.150, 95% CI: 1.021-1.296, P = 0.021) and interleukin-1beta (IL-1β) (OR = 1.152, 95% CI: 1.003-1.325, P = 0.046) were positively associated with lung adenocarcinoma risk, though no inflammatory factors showed causal links to squamous cell lung cancer risk. Stratified by smoking status, interferon gamma-induced protein 10 (IP-10) (OR = 0.861, 95% CI: 0.781-0.950, P = 0.003) was inversely associated while IL-1β (OR = 1.190, 95% CI: 1.023-1.384, P = 0.024) was positively associated with lung cancer risk in ever smokers. Among never smokers, a positive association was observed between lung cancer risk and SCF (OR = 1.474, 95% CI: 1.105-1.964, P = 0.008). Importantly, these causal inferences remained robust across multiple complementary MR approaches, including MR-Egger, weighted median, weighted mode and simple mode regressions. Sensitivity analyses also excluded potential bias stemming from pleiotropy. Conclusion This MR study found preliminary evidence that genetically predicted levels of four inflammatory cytokines-SCF, IL-1β, IL-18, and IP-10-may causally influence lung cancer risk in an overall and subtype-specific manner, as well as stratified by smoking status. Identifying these cytokine pathways that may promote lung carcinogenesis represents potential new targets for the prevention, early detection, and treatment of this deadly malignancy.
Collapse
Affiliation(s)
- Dachen Luo
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zonglian Gong
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qingyuan Zhan
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Center for Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Shan Lin
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
119
|
Ferrucci V, Miceli M, Pagliuca C, Bianco O, Castaldo L, Izzo L, Cozzolino M, Zannella C, Oglio F, Polcaro A, Randazzo A, Colicchio R, Galdiero M, Berni Canani R, Salvatore P, Zollo M. Modulation of innate immunity related genes resulting in prophylactic antimicrobial and antiviral properties. J Transl Med 2024; 22:574. [PMID: 38886736 PMCID: PMC11184722 DOI: 10.1186/s12967-024-05378-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND The innate immunity acts during the early phases of infection and its failure in response to a multilayer network of co-infections is cause of immune system dysregulation. Epidemiological SARS-CoV-2 infections data, show that Influenza Virus (FLU-A-B-C) and Respiratory Syncytial Virus (RSV) are co-habiting those respiratory traits. These viruses, especially in children (mostly affected by 'multi-system inflammatory syndrome in children' [MIS-C] and the winter pandemic FLU), in the aged population, and in 'fragile' patients are causing alteration in immune response. Then, bacterial and fungal pathogens are also co-habiting the upper respiratory traits (e.g., Staphylococcus aureus and Candida albicans), thus contributing to morbidity in those COVID-19 affected patients. METHODS Liquid chromatography coupled with high-resolution mass spectrometry using the quadrupole orbital ion trap analyser (i.e., UHPLC-Q-Orbitrap HRMS) was adopted to measure the polyphenols content of a new nutraceutical formula (Solution-3). Viral infections with SARS-CoV-2 (EG.5), FLU-A and RSV-A viruses (as performed in BLS3 authorised laboratory) and real time RT-PCR (qPCR) assay were used to test the antiviral action of the nutraceutical formula. Dilution susceptibility tests have been used to estimate the minimum inhibitory and bactericidal concentration (MIC and MBC, respectively) of Solution-3 on a variety of microorganisms belonging to Gram positive/ negative bacteria and fungi. Transcriptomic data analyses and functional genomics (i.e., RNAseq and data mining), coupled to qPCR and ELISA assays have been used to investigate the mechanisms of action of the nutraceutical formula on those processes involved in innate immune response. RESULTS Here, we have tested the combination of natural products containing higher amounts of polyphenols (i.e., propolis, Verbascum thapsus L., and Thymus vulgaris L.), together with the inorganic long chain polyphosphates 'polyPs' with antiviral, antibacterial, and antifungal behaviours, against SARS-CoV-2, FLU-A, RSV-A, Gram positive/ negative bacteria and fungi (i.e., Candida albicans). These components synergistically exert an immunomodulatory action by enhancing those processes involved in innate immune response (e.g., cytokines: IFNγ, TNFα, IL-10, IL-6/12; chemokines: CXCL1; antimicrobial peptides: HBD-2, LL-37; complement system: C3). CONCLUSION The prophylactic antimicrobial success of this nutraceutical formula against SARS-CoV-2, FLU-A and RSV-A viruses, together with the common bacteria and fungi co-infections as present in human oral cavity, is expected to be valuable.
Collapse
Affiliation(s)
- Veronica Ferrucci
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
| | - Marco Miceli
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Chiara Pagliuca
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Orazio Bianco
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Luigi Castaldo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Luana Izzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Marica Cozzolino
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Franca Oglio
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Antonio Polcaro
- Polcaro Fitopreparazioni S.R.L, Via Sant Agnello, 9 D; 80030, Roccarainola, Naples, Italy
| | - Antonio Randazzo
- Department of Pharmacy, University of Naples 'Federico II', Via Domenico Montesano 49, 80131, Naples, Italy
| | - Roberta Colicchio
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- UOC of Virology and Microbiology, University Hospital of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Roberto Berni Canani
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
- Dipartimento Di Scienze Mediche Traslazionali, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Paola Salvatore
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy
| | - Massimo Zollo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples 'Federico II', Via Sergio Pansini 5, 80131, Naples, Italy.
- CEINGE Biotecnologie Avanzate 'Franco Salvatore', Via Gaetano Salvatore 486, 80145, Naples, Italy.
- Elysium Cell Bio Ita, Via Gaetano Salvatore 486, 80145, Naples, Italy.
- DAI Medicina di Laboratorio e Trasfusionale, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
120
|
Lian GY, Wang QM, Mak TSK, Huang XR, Yu XQ, Lan HY. Disrupting Smad3 potentiates immunostimulatory function of NK cells against lung carcinoma by promoting GM-CSF production. Cell Mol Life Sci 2024; 81:262. [PMID: 38878186 PMCID: PMC11335298 DOI: 10.1007/s00018-024-05290-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 04/28/2024] [Accepted: 05/23/2024] [Indexed: 06/29/2024]
Abstract
Through Smad3-dependent signalings, transforming growth factor-β (TGF-β) suppresses the development, maturation, cytokine productions and cytolytic functions of NK cells in cancer. Silencing Smad3 remarkably restores the cytotoxicity of NK-92 against cancer in TGF-β-rich microenvironment, but its effects on the immunoregulatory functions of NK cells remain obscure. In this study, we identified Smad3 functioned as a transcriptional repressor for CSF2 (GM-CSF) in NK cells. Therefore, disrupting Smad3 largely mitigated TGF-β-mediated suppression on GM-CSF production by NK cells. Furthermore, silencing GM-CSF in Smad3 knockout NK cells substantially impaired their anti-lung carcinoma effects. In-depth study demonstrated that NK-derived GM-CSF strengthened T cell immune responses by stimulating dendritic cell differentiation and M1 macrophage polarization. Meanwhile, NK-derived GM-CSF promoted the survival of neutrophils, which in turn facilitated the terminal maturation of NK cells, and subsequently boosted NK-cell mediated cytotoxicity against lung carcinoma. Thus, Smad3-silenced NK-92 (NK-92-S3KD) may serve as a promising immunoadjuvant therapy with clinical translational value given its robust cytotoxicity against malignant cells and immunostimulatory functions to reinforce the therapeutic effects of other immunotherapies.
Collapse
Affiliation(s)
- Guang-Yu Lian
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing-Ming Wang
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Thomas Shiu-Kwong Mak
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ru Huang
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xue-Qing Yu
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Hui-Yao Lan
- Guangdong-Hong Kong Joint Research Laboratory on Immunological and Genetic Kidney Diseases, Departments of Pathology and Nephrology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
- Department of Medicine & Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
121
|
Dutta S, Mudaranthakam DP, Li Y, Sardiu ME. PerSEveML: a web-based tool to identify persistent biomarker structure for rare events using an integrative machine learning approach. Mol Omics 2024; 20:348-358. [PMID: 38690925 DOI: 10.1039/d4mo00008k] [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: 05/03/2024]
Abstract
Omics data sets often pose a computational challenge due to their high dimensionality, large size, and non-linear structures. Analyzing these data sets becomes especially daunting in the presence of rare events. Machine learning (ML) methods have gained traction for analyzing rare events, yet there has been limited exploration of bioinformatics tools that integrate ML techniques to comprehend the underlying biology. Expanding upon our previously developed computational framework of an integrative machine learning approach, we introduce PerSEveML, an interactive web-based tool that uses crowd-sourced intelligence to predict rare events and determine feature selection structures. PerSEveML provides a comprehensive overview of the integrative approach through evaluation metrics that help users understand the contribution of individual ML methods to the prediction process. Additionally, PerSEveML calculates entropy and rank scores, which visually organize input features into a persistent structure of selected, unselected, and fluctuating categories that help researchers uncover meaningful hypotheses regarding the underlying biology. We have evaluated PerSEveML on three diverse biologically complex data sets with extremely rare events from small to large scale and have demonstrated its ability to generate valid hypotheses. PerSEveML is available at https://biostats-shinyr.kumc.edu/PerSEveML/ and https://github.com/sreejatadutta/PerSEveML.
Collapse
Affiliation(s)
- Sreejata Dutta
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
| | - Yanming Li
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
| | - Mihaela E Sardiu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
122
|
Hurme P, Kähkönen M, Rückert B, Vahlberg T, Turunen R, Vuorinen T, Akdis M, Akdis CA, Jartti T. Disease Severity and Cytokine Expression in the Rhinovirus-Induced First Wheezing Episode. Viruses 2024; 16:924. [PMID: 38932217 PMCID: PMC11209381 DOI: 10.3390/v16060924] [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: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Wheezing children infected with rhinovirus (RV) have a markedly increased risk of subsequently developing recurrencies and asthma. No previous studies have assessed the association between cytokine response and the severity of acute illness in the first wheezing episode in children infected with RV. Forty-seven children treated both as inpatients and as outpatients infected with RV only, aged 3-23 months, with severe first wheezing episodes were recruited. During acute illness, peripheral blood mononuclear cells (PBMCs) were isolated and stimulated with anti-CD3/anti-CD28 in vitro. A multiplex ELISA was used to quantitatively identify 56 different cytokines. The mean age of the children was 17 months, 74% were males, 79% were hospitalized, and 33% were sensitized. In adjusted analyses, the inpatient group was characterized by decreased expressions of interferon gamma (IFN-γ), interleukin 10 (IL-10), macrophage inflammatory protein 1 alpha (MIP-1α), RANTES (CCL5), and tumor necrosis factor-alpha (TNF-α) and an increased expression of ENA-78 (CXCL5) compared to the outpatient group. The cytokine response profiles from the PBMCs were different between the inpatient and outpatient groups. Our results support that firmly controlled interplay between pro-inflammatory and anti-inflammatory responses are required during acute viral infection to absolve the initial infection leading, to less severe illness.
Collapse
Affiliation(s)
- Pekka Hurme
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, 20520 Turku, Finland
| | - Miisa Kähkönen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, 20520 Turku, Finland
| | - Beate Rückert
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Christine Kühne-Center for Allergy Research and Education (CK-CARE), 7265 Davos, Switzerland
| | - Tero Vahlberg
- Department of Biostatistics, Turku University Hospital and University of Turku, 20520 Turku, Finland
| | - Riitta Turunen
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, 20520 Turku, Finland
- New Children’s Hospital, Helsinki University Hospital and University of Helsinki, 00290 Helsinki, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku and Turku University Hospital, 20520 Turku, Finland
- Department of Clinical Microbiology, Turku University Hospital, 20520 Turku, Finland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Christine Kühne-Center for Allergy Research and Education (CK-CARE), 7265 Davos, Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zürich, Christine Kühne-Center for Allergy Research and Education (CK-CARE), 7265 Davos, Switzerland
| | - Tuomas Jartti
- Department of Pediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, 20520 Turku, Finland
| |
Collapse
|
123
|
Cunha D, Neves M, Silva D, Silvestre AR, Nunes PB, Arrobas F, Ribot JC, Ferreira F, Moita LF, Soares-de-Almeida L, Silva JM, Filipe P, Ferreira J. Tumor-Infiltrating T Cells in Skin Basal Cell Carcinomas and Squamous Cell Carcinomas: Global Th1 Preponderance with Th17 Enrichment-A Cross-Sectional Study. Cells 2024; 13:964. [PMID: 38891095 PMCID: PMC11172364 DOI: 10.3390/cells13110964] [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: 05/04/2024] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
Basal cell carcinomas (BCCs) and squamous cell carcinomas (SCCs) are high-incidence, non-melanoma skin cancers (NMSCs). The success of immune-targeted therapies in advanced NMSCs led us to anticipate that NMSCs harbored significant populations of tumor-infiltrating lymphocytes with potential anti-tumor activity. The main aim of this study was to characterize T cells infiltrating NMSCs. Flow cytometry and immunohistochemistry were used to assess, respectively, the proportions and densities of T cell subpopulations in BCCs (n = 118), SCCs (n = 33), and normal skin (NS, n = 30). CD8+ T cells, CD4+ T cell subsets, namely, Th1, Th2, Th17, Th9, and regulatory T cells (Tregs), CD8+ and CD4+ memory T cells, and γδ T cells were compared between NMSCs and NS samples. Remarkably, both BCCs and SCCs featured a significantly higher Th1/Th2 ratio (~four-fold) and an enrichment for Th17 cells. NMSCs also showed a significant enrichment for IFN-γ-producing CD8+T cells, and a depletion of γδ T cells. Using immunohistochemistry, NMSCs featured denser T cell infiltrates (CD4+, CD8+, and Tregs) than NS. Overall, these data favor a Th1-predominant response in BCCs and SCCs, providing support for immune-based treatments in NMSCs. Th17-mediated inflammation may play a role in the progression of NMSCs and thus become a potential therapeutic target in NMSCs.
Collapse
Affiliation(s)
- Daniela Cunha
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Centro de Dermatologia, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal
- Dermatology Unit, Champalimaud Foundation, 1400-038 Lisbon, Portugal
| | - Marco Neves
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Daniela Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Ana Rita Silvestre
- Serviço de Anatomia Patológica, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal (P.B.N.)
| | - Paula Borralho Nunes
- Serviço de Anatomia Patológica, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal (P.B.N.)
- Instituto de Anatomia Patológica, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Fernando Arrobas
- Datamedica, Biostatistics Services and Consulting, 2610-008 Amadora, Portugal
| | - Julie C. Ribot
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
| | - Fernando Ferreira
- CIISA—Centre for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, 1300-477 Lisbon, Portugal;
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Luís F. Moita
- Innate Immunity and Inflammation Laboratory, Instituto Gulbenkian de Ciência, 2780-156 Oeiras, Portugal
| | - Luís Soares-de-Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - João Maia Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Centro de Dermatologia, Hospital CUF Descobertas, 1998-018 Lisbon, Portugal
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Paulo Filipe
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Serviço de Dermatologia, Centro Hospitalar Universitário Lisboa Norte EPE, 1649-028 Lisbon, Portugal
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - João Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal; (D.C.)
- Clínica Dermatológica Universitária, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal
| |
Collapse
|
124
|
Chen C, Zhou S, Yang X, Ren M, Qi Y, Mao Y, Yang C. In vitro study of cold atmospheric plasma-activated liquids inhibits malignant melanoma by affecting macrophage polarization through the ROS/JAK2/STAT1 pathway. Biomed Pharmacother 2024; 175:116657. [PMID: 38688171 DOI: 10.1016/j.biopha.2024.116657] [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: 01/21/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024] Open
Abstract
Melanoma is a prevalent malignant skin tumor known for its high invasive ability and a high rate of metastasis, making clinical treatment exceptionally challenging. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment and play a crucial role in tumor survival and development. Cold atmospheric plasma (CAP) is an emerging tool for tumor treatment that has garnered attention from scholars due to its interaction with non-tumor cells in the tumor microenvironment. Here, we used the macrophage lines THP-1 and RAW264.7, as well as the melanoma cell lines A375 and MV3, as research subjects to investigate the effect of plasma-activated liquid (PAL) on macrophage differentiation and its inhibitory effect on melanoma cell proliferation. We confirmed that the killing effect of PAL on melanoma cells was selective. Using flow cytometry and PCR, we discovered that PAL can influence macrophage differentiation. Through in vitro cell coculture, we demonstrated that PAL-treated macrophages can significantly impede tumor cell development and progression, and the effect is more potent than that of PAL directly targeting tumor cells. Furthermore, we have proposed the hypothesis that PAL promotes the differentiation of macrophages into the M1 type through the ROS/JAK2/STAT1 pathway. To test the hypothesis, we employed catalase and fludarabine to block different sites of the pathway. The results were then validated through Western Blot, qPCR and ELISA. This study illustrates that PAL therapy is an effective tumor immunotherapy and expands the scope of tumor immunotherapy. Furthermore, these findings establish a theoretical foundation for potential clinical applications of PAL.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Shiyun Zhou
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Xingyu Yang
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Miaomiao Ren
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Yongshuang Qi
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Yiwen Mao
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China
| | - Chunjun Yang
- Department of Dermatology and Venereology, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, the Second Affiliated Hospital of Anhui Medical University, Anhui medical University, Hefei 230601, China.
| |
Collapse
|
125
|
Shi J, Wu W, Chen D, Liao Z, Sheng T, Wang Y, Yao Y, Wu Q, Liu F, Zhou R, Zhu C, Shen X, Mao Z, Ding Y, Wang W, Dotti G, Sun J, Liang X, Fang W, Zhao P, Li H, Gu Z. Lyophilized lymph nodes for improved delivery of chimeric antigen receptor T cells. NATURE MATERIALS 2024; 23:844-853. [PMID: 38448658 DOI: 10.1038/s41563-024-01825-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2024] [Indexed: 03/08/2024]
Abstract
Lymph nodes are crucial organs of the adaptive immune system, orchestrating T cell priming, activation and tolerance. T cell activity and function are highly regulated by lymph nodes, which have a unique structure harbouring distinct cells that work together to detect and respond to pathogen-derived antigens. Here we show that implanted patient-derived freeze-dried lymph nodes loaded with chimeric antigen receptor T cells improve delivery to solid tumours and inhibit tumour recurrence after surgery. Chimeric antigen receptor T cells can be effectively loaded into lyophilized lymph nodes, whose unaltered meshwork and cytokine and chemokine contents promote chimeric antigen receptor T cell viability and activation. In mouse models of cell-line-derived human cervical cancer and patient-derived pancreatic cancer, delivery of chimeric antigen receptor T cells targeting mesothelin via the freeze-dried lymph nodes is more effective in preventing tumour recurrence when compared to hydrogels containing T-cell-supporting cytokines. This tissue-mediated cell delivery strategy holds promise for controlled release of various cells and therapeutics with long-term activity and augmented function.
Collapse
Affiliation(s)
- Jiaqi Shi
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Wei Wu
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dong Chen
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ziyan Liao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Tao Sheng
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yanfang Wang
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuejun Yao
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qing Wu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Feng Liu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Ruyi Zhou
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chaojie Zhu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinyuan Shen
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yuan Ding
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weilin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jie Sun
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Xiao Liang
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weijia Fang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hongjun Li
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute, Zhejiang University, Jinhua, China.
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Jinhua Institute, Zhejiang University, Jinhua, China.
- Key Laboratory for Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
126
|
Xiao S, Ma S, Sun B, Pu W, Duan S, Han J, Hong Y, Zhang J, Peng Y, He C, Yi P, Caligiuri MA, Yu J. The tumor-intrinsic role of the m 6A reader YTHDF2 in regulating immune evasion. Sci Immunol 2024; 9:eadl2171. [PMID: 38820140 DOI: 10.1126/sciimmunol.adl2171] [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: 10/05/2023] [Accepted: 05/09/2024] [Indexed: 06/02/2024]
Abstract
Tumors evade attacks from the immune system through various mechanisms. Here, we identify a component of tumor immune evasion mediated by YTH domain-containing family protein 2 (YTHDF2), a reader protein that usually destabilizes m6A-modified mRNA. Loss of tumoral YTHDF2 inhibits tumor growth and prolongs survival in immunocompetent tumor models. Mechanistically, tumoral YTHDF2 deficiency promotes the recruitment of macrophages via CX3CL1 and enhances mitochondrial respiration of CD8+ T cells by impairing tumor glycolysis metabolism. Tumoral YTHDF2 deficiency promotes inflammatory macrophage polarization and antigen presentation in the presence of IFN-γ. In addition, IFN-γ induces autophagic degradation of tumoral YTHDF2, thereby sensitizing tumor cells to CD8+ T cell-mediated cytotoxicity. Last, we identified a small molecule compound that preferentially induces YTHDF2 degradation, which shows a potent antitumor effect alone but a better effect when combined with anti-PD-L1 or anti-PD-1 antibodies. Collectively, YTHDF2 appears to be a tumor-intrinsic regulator that orchestrates immune evasion, representing a promising target for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Sai Xiao
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
| | - Baofa Sun
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wenchen Pu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, China
| | - Songqi Duan
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China
| | - Jingjing Han
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Yaqun Hong
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA 91010, USA
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, China
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL 60637, USA
| | - Ping Yi
- Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA 91010, USA
- Comprehensive Cancer Center, City of Hope, Los Angeles, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Los Angeles, CA 91010, USA
| |
Collapse
|
127
|
Qiu X, Nair MG, Jaroszewski L, Godzik A. Deciphering Abnormal Platelet Subpopulations in COVID-19, Sepsis and Systemic Lupus Erythematosus through Machine Learning and Single-Cell Transcriptomics. Int J Mol Sci 2024; 25:5941. [PMID: 38892129 PMCID: PMC11173046 DOI: 10.3390/ijms25115941] [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: 04/16/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This study focuses on understanding the transcriptional heterogeneity of activated platelets and its impact on diseases such as sepsis, COVID-19, and systemic lupus erythematosus (SLE). Recognizing the limited knowledge in this area, our research aims to dissect the complex transcriptional profiles of activated platelets to aid in developing targeted therapies for abnormal and pathogenic platelet subtypes. We analyzed single-cell transcriptional profiles from 47,977 platelets derived from 413 samples of patients with these diseases, utilizing Deep Neural Network (DNN) and eXtreme Gradient Boosting (XGB) to distinguish transcriptomic signatures predictive of fatal or survival outcomes. Our approach included source data annotations and platelet markers, along with SingleR and Seurat for comprehensive profiling. Additionally, we employed Uniform Manifold Approximation and Projection (UMAP) for effective dimensionality reduction and visualization, aiding in the identification of various platelet subtypes and their relation to disease severity and patient outcomes. Our results highlighted distinct platelet subpopulations that correlate with disease severity, revealing that changes in platelet transcription patterns can intensify endotheliopathy, increasing the risk of coagulation in fatal cases. Moreover, these changes may impact lymphocyte function, indicating a more extensive role for platelets in inflammatory and immune responses. This study identifies crucial biomarkers of platelet heterogeneity in serious health conditions, paving the way for innovative therapeutic approaches targeting platelet activation, which could improve patient outcomes in diseases characterized by altered platelet function.
Collapse
Affiliation(s)
| | | | | | - Adam Godzik
- Division of Biomedical Sciences, University of California Riverside School of Medicine, Riverside, CA 92521, USA; (X.Q.); (M.G.N.); (L.J.)
| |
Collapse
|
128
|
Wang J, Lou Y, Wang S, Zhang Z, You J, Zhu Y, Yao Y, Hao Y, Liu P, Xu LX. IFNγ at the early stage induced after cryo-thermal therapy maintains CD4 + Th1-prone differentiation, leading to long-term antitumor immunity. Front Immunol 2024; 15:1345046. [PMID: 38827732 PMCID: PMC11140566 DOI: 10.3389/fimmu.2024.1345046] [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: 11/27/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction Recently, more and more research illustrated the importance of inducing CD4+ T helper type (Th)-1 dominant immunity for the success of tumor immunotherapy. Our prior studies revealed the crucial role of CD4+ Th1 cells in orchestrating systemic and durable antitumor immunity, which contributes to the satisfactory outcomes of the novel cryo-thermal therapy in the B16F10 tumor model. However, the mechanism for maintaining the cryo-thermal therapy-mediated durable CD4+ Th1-dominant response remains uncovered. Additionally, cryo-thermal-induced early-stage CD4+ Th1-dominant T cell response showed a correlation with the favorable prognosis in patients with colorectal cancer liver metastasis (CRCLM). We hypothesized that CD4+ Th1-dominant differentiation induced during the early stage post cryo-thermal therapy would affect the balance of CD4+ subsets at the late phase. Methods To understand the role of interferon (IFN)-γ, the major effector of Th1 subsets, in maintaining long-term CD4+ Th1-prone polarization, B16F10 melanoma model was established in this study and a monoclonal antibody was used at the early stage post cryo-thermal therapy for interferon (IFN)-γ signaling blockade, and the influence on the phenotypic and functional change of immune cells was evaluated. Results IFNγ at the early stage after cryo-thermal therapy maintained long-lasting CD4+ Th1-prone immunity by directly controlling Th17, Tfh, and Tregs polarization, leading to the hyperactivation of Myeloid-derived suppressor cells (MDSCs) represented by abundant interleukin (IL)-1β generation, and thereby further amplifying Th1 response. Discussion Our finding emphasized the key role of early-phase IFNγ abundance post cryo-thermal therapy, which could be a biomarker for better prognosis after cryo-thermal therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping Liu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lisa X. Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
129
|
Li T, Yi J, Wu H, Wang K, Zhou B. SLC7A11 in hepatocellular carcinoma: potential mechanisms, regulation, and clinical significance. Am J Cancer Res 2024; 14:2326-2342. [PMID: 38859833 PMCID: PMC11162675 DOI: 10.62347/kgcl7357] [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: 03/06/2024] [Accepted: 05/13/2024] [Indexed: 06/12/2024] Open
Abstract
Exploring novel early detection biomarkers and developing more efficacious treatments remain pressing tasks in the current research landscape for hepatocellular carcinoma (HCC). Morphologically and molecularly separate from apoptosis, cell death, and autophagy, ferroptosis is a recently discovered, unique, controlled form of cell death. SLC7A11 (also known as xCT) represents a subunit of the cystine-glutamate antiporter (also known as system Xc(-)). A growing body of research suggests that induction of ferroptosis through SLC7A11 can effectively eliminate hepatocellular carcinoma (HCC) cells, particularly those exhibiting resistance to alternative forms of cell death. Thus, targeting ferroptosis via SLC7A11 may become a new direction for the design of therapeutic strategies for HCC. Although many research articles have investigated the possible roles of SLC7A11 in HCC, a study that summarizes the main findings, including the regulators and mechanisms of action of SLC7A11 in HCC is not available. Therefore, we present a comprehensive overview of the functions of ferroptosis, particularly SLC7A11, in the identification, development, and management of HCC in this review. In addition, we discuss how this knowledge can be translated into treatment by providing a systemic therapy in advanced HCC using sorafenib, the first-line drug targeting multiple kinases and SLC7A11. We further dissect the possible barriers as well as the corresponding solutions and provide insights on how to navigate effective treatment using this knowledge.
Collapse
Affiliation(s)
- Tianze Li
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
- Queen Mary School, Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| | - Jianwei Yi
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| | - Huajun Wu
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| | - Kai Wang
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
- Jiangxi Province Engineering Research Center of Hepatobiliary DiseaseNanchang 330006, Jiangxi, P. R. China
| | - Binghai Zhou
- Division of Hepato-Biliary-Pancreatic Surgery, Department of General Surgery, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, Jiangxi, P. R. China
| |
Collapse
|
130
|
Hu YM, Zhao F, Graff JN, Chen C, Zhao X, Thomas GV, Wu H, Kardosh A, Mills GB, Alumkal JJ, Moran AE, Xia Z. Androgen receptor activity inversely correlates with immune cell infiltration and immunotherapy response across multiple cancer lineages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593181. [PMID: 38798471 PMCID: PMC11118439 DOI: 10.1101/2024.05.08.593181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
There is now increasing recognition of the important role of androgen receptor (AR) in modulating immune function. To gain a comprehensive understanding of the effects of AR activity on cancer immunity, we employed a computational approach to profile AR activity in 33 human tumor types using RNA-Seq datasets from The Cancer Genome Atlas. Our pan-cancer analysis revealed that the genes most negatively correlated with AR activity across cancers are involved in active immune system processes. Importantly, we observed a significant negative correlation between AR activity and IFNγ pathway activity at the pan-cancer level. Indeed, using a matched biopsy dataset from subjects with prostate cancer before and after AR-targeted treatment, we verified that inhibiting AR enriches immune cell abundances and is associated with higher IFNγ pathway activity. Furthermore, by analyzing immunotherapy datasets in multiple cancers, our results demonstrate that low AR activity was significantly associated with a favorable response to immunotherapy. Together, our data provide a comprehensive assessment of the relationship between AR signaling and tumor immunity.
Collapse
Affiliation(s)
- Ya-Mei Hu
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Faming Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Julie N. Graff
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- VA Portland Health Care System, Portland, OR, USA
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Xiyue Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - George V. Thomas
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Pathology & Laboratory Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Hui Wu
- Division of Biomaterial and Biomedical Sciences, Department of Oral Rehabilitation and Biosciences, Oregon Health & Science University, Portland, OR, USA
| | - Adel Kardosh
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Gordon B. Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Joshi J. Alumkal
- Department of Internal Medicine, Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Amy E. Moran
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Center for Biomedical Data Science, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
131
|
Chen X. From immune equilibrium to tumor ecodynamics. Front Oncol 2024; 14:1335533. [PMID: 38807760 PMCID: PMC11131381 DOI: 10.3389/fonc.2024.1335533] [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: 11/15/2023] [Accepted: 04/01/2024] [Indexed: 05/30/2024] Open
Abstract
Objectives There is no theory to quantitatively describe the complex tumor ecosystem. At the same time, cancer immunotherapy is considered a revolution in oncology, but the methods used to describe tumors and the criteria used to evaluate efficacy are not keeping pace. The purpose of this study is to establish a new theory for quantitatively describing the tumor ecosystem, innovating the methods of tumor characterization, and establishing new efficacy evaluation criteria for cancer immunotherapy. Methods Based on the mathematization of immune equilibrium theory and the establishment of immunodynamics in a previous study, the method of reverse immunodynamics was used, namely, the immune braking force was regarded as the tumor ecological force and the immune force was regarded as the tumor ecological braking force, and the concept of momentum in physics was applied to the tumor ecosystem to establish a series of tumor ecodynamic equations. These equations were used to solve the fundamental and applied problems of the complex tumor ecosystem. Results A series of tumor ecodynamic equations were established. The tumor ecological momentum equations and their component factors could be used to distinguish disease progression, pseudoprogression, and hyperprogression in cancer immunotherapy. On this basis, the adjusted tumor momentum equations were established to achieve the equivalence of tumor activity (including immunosuppressive activity and metabolic activity) and tumor volume, which could be used to calculate individual disease remission rate and establish new efficacy evaluation criteria (ieRECIST) for immunotherapy of solid tumor based on tumor ecodynamics. At the same time, the concept of moving cube-to-force square ratio and its expression were proposed to calculate the area under the curve of tumor ecological braking force of blood required to achieve an individual disease remission rate when the adjusted tumor ecological momentum was known. Conclusions A new theory termed tumor ecodynamics emphasizing both tumor activity and tumor volume is established to solve a series of basic and applied problems in the complex tumor ecosystem. It can be predicted that the future will be the era of cancer immune ecotherapy that targets the entire tumor ecosystem.
Collapse
Affiliation(s)
- Xiaoping Chen
- State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- CAS Lamvac (Guangzhou) Biomedical Technology Co., Ltd., Guangzhou, China
| |
Collapse
|
132
|
da Silva MI, Oli N, Gambonini F, Ott T. Effects of parity and early pregnancy on peripheral blood leukocytes in dairy cattle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592827. [PMID: 38766084 PMCID: PMC11100682 DOI: 10.1101/2024.05.06.592827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Subfertility remains a major problem in the dairy industry. Only 35-40% of high-yielding dairy cows and 55-65% of nonlactating heifers become pregnant after their first service. The immune system plays a critical role in the establishment of pregnancy. However, it can also create challenges for embryo survival and contribute to reduced fertility. We conducted 2 separate experiments to characterize changes in subsets of peripheral blood leukocytes (PBL) and their phenotype over the estrous cycle and early pregnancy in heifers and cows. We used flow cytometry and RT-qPCR to assess protein and mRNA expression of molecules important for immune function. We observed that monocytes and T cells were most affected by pregnancy status in heifers, whereas, CD8+ lymphocytes and natural killer (NK) cells were most affected during early pregnancy in cows. Changes in immune parameters measured appeared to be greater in heifers than cows including changes in expression of numerous immune function molecules. To test the hypothesis, we conducted a third experiment to simultaneously analyze the immunological responses to pregnancy between cows and heifers. We observed that cows had greater expression of proinflammatory cytokines and molecules associated with leukocyte migration and phagocytosis compared to heifers. Moreover, animals that failed to become pregnant showed altered expression of anti-inflammatory molecules. Overall, these findings support the hypothesis that early pregnancy signaling alters the proportions and functions of peripheral blood immune cells and differences between cows and heifers may yield insight into the reduced fertility of mature lactating dairy cows.
Collapse
Affiliation(s)
- M I da Silva
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - N Oli
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - F Gambonini
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - T Ott
- Department of Animal Science, Center for Reproductive Biology and Health, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
133
|
Das R, Mishra P, Mishra B, Jha R. Effect of in ovo feeding of xylobiose and xylotriose on plasma immunoglobulin, cecal metabolites production, microbial ecology, and metabolic pathways in broiler chickens. J Anim Sci Biotechnol 2024; 15:62. [PMID: 38702804 PMCID: PMC11069197 DOI: 10.1186/s40104-024-01022-7] [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: 11/16/2023] [Accepted: 03/06/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Dietary supplementation of xylooligosaccharides (XOS) has been found to influence gut health by manipulating cecal microbiota and producing microbe-origin metabolites. But no study investigated and compared the effect of in ovo feeding of xylobiose (XOS2) and xylotriose (XOS3) in chickens. This study investigated the effect of in ovo feeding of these XOS compounds on post-hatch gut health parameters in chickens. A total of 144 fertilized chicken eggs were divided into three groups: a) non-injected control (CON), b) XOS2, and c) XOS3. On the 17th embryonic day, the eggs of the XOS2 and XOS3 groups were injected with 3 mg of XOS2 and XOS3 diluted in 0.5 mL of 0.85% normal saline through the amniotic sac. After hatching, the chicks were raised for 21 d. Blood was collected on d 14 to measure plasma immunoglobulin. Cecal digesta were collected for measuring short-chain fatty acids (SCFA) on d 14 and 21, and for microbial ecology and microbial metabolic pathway analyses on d 7 and 21. RESULTS The results were considered significantly different at P < 0.05. ELISA quantified plasma IgA and IgG on d 14 chickens, revealing no differences among the treatments. Gas chromatography results showed no significant differences in the concentrations of cecal SCFAs on d 14 but significant differences on d 21. However, the SCFA concentrations were lower in the XOS3 than in the CON group on d 21. The cecal metagenomics data showed that the abundance of the family Clostridiaceae significantly decreased on d 7, and the abundance of the family Oscillospiraceae increased on d 21 in the XOS2 compared to the CON. There was a reduction in the relative abundance of genus Clostridium sensu stricto 1 in the XOS2 compared to the CON on d 7 and the genus Ruminococcus torques in both XOS2 and XOS3 groups compared to the CON on d 21. The XOS2 and XOS3 groups reduced the genes for chondroitin sulfate degradation I and L-histidine degradation I pathways, which contribute to improved gut health, respectively, in the microbiome on d 7. In contrast, on d 21, the XOS2 and XOS3 groups enriched the thiamin salvage II, L-isoleucine biosynthesis IV, and O-antigen building blocks biosynthesis (E. coli) pathways, which are indicative of improved gut health. Unlike the XOS3 and CON, the microbiome enriched the pathways associated with energy enhancement, including flavin biosynthesis I, sucrose degradation III, and Calvin-Benson-Bassham cycle pathways, in the XOS2 group on d 21. CONCLUSION In ovo XOS2 and XOS3 feeding promoted beneficial bacterial growth and reduced harmful bacteria at the family and genus levels. The metagenomic-based microbial metabolic pathway profiling predicted a favorable change in the availability of cecal metabolites in the XOS2 and XOS3 groups. The modulation of microbiota and metabolic pathways suggests that in ovo XOS2 and XOS3 feeding improved gut health during the post-hatch period of broilers.
Collapse
Affiliation(s)
- Razib Das
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Pravin Mishra
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Birendra Mishra
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, HI, 96822, USA.
| |
Collapse
|
134
|
Balan Y, Sundaramurthy R, Gaur A, Varatharajan S, Raj GM. Impact of high-salt diet in health and diseases and its role in pursuit of cancer immunotherapy by modulating gut microbiome. J Family Med Prim Care 2024; 13:1628-1635. [PMID: 38948582 PMCID: PMC11213449 DOI: 10.4103/jfmpc.jfmpc_1574_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 07/02/2024] Open
Abstract
Cancer chemotherapy remains an area of concern, as many of the therapies are uncomfortable involving side effects and unpleasant experiences. These factors could further reduce patient's quality of life, and even endanger their life. Many therapeutic strategies have been tried to reduce the unpleasant side effects and increase the treatment effectiveness; however, none have shown to have promising effects. One of the main hindrances to cancer therapy is the escape strategies by tumor cells to the immune attack. Promoting inflammation in the tumor microenvironment is the cornerstone and key therapeutic target in cancer chemotherapy. High-salt diet (HSD) intake, though it has deleterious effects on human health by promoting chronic inflammation, is found to be advantageous in the tumor microenvironment. Studies identified HSD favors an increased abundance of Bifidobacterium species in the tumor environment due to gut barrier alteration, which, in turn, promotes inflammation and favors improved response to cancer chemotherapy. A review of the literature was carried out to find out the effects of an HSD on health and diseases, with special mention of its effect on cancer chemotherapy. Studies emphasized HSD would block the myeloid-derived suppressor cells which will enhance the tumor immunity. Exploration of the precise mechanism of simple HSD regime/ingestion of specific bacterial species as probiotics will be effective and essential to formulate the game-changing cancer chemotherapy. With the modern era of healthcare moving toward precision medicine where the physician can choose the treatment option suitable for the individual, HSD regime/ingestion of specific bacterial species can be considered.
Collapse
Affiliation(s)
- Yuvaraj Balan
- Department of Biochemistry, All India Institute of Medical Sciences, Madurai, Tamil Nadu, India
| | - Raja Sundaramurthy
- Department of Microbiology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Archana Gaur
- Department of Physiology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| | | | - Gerard Marshall Raj
- Department of Pharmacology, All India Institute of Medical Sciences, Hyderabad, Telangana, India
| |
Collapse
|
135
|
Abd El Rahiem RA, Ibrahim SA, Effat H, El-Houseini ME, Osman RA, Abdelraouf A, Elzayat EM. Curcumin, Piperine and Taurine Combination Enhances the Efficacy of Transarterial Chemoembolization Therapy in patients with Intermediate Stage Hepatocellular Carcinoma: A Pilot Study. Asian Pac J Cancer Prev 2024; 25:1589-1598. [PMID: 38809630 PMCID: PMC11318834 DOI: 10.31557/apjcp.2024.25.5.1589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2024] Open
Abstract
INTRODUCTION Diagnosis of the majority of hepatocellular carcinoma (HCC) patients occurs at intermediate to advanced stages, with a few curative therapeutic options being available. It is therefore strongly urgent to discover additional adjuvant therapy for this lethal malignancy. This study aimed to assess the effectiveness of curcumin (C), piperine (P) and taurine (T) combination as adjuvant agents on serum levels of IFN-γ, immunophenotypic and molecular characterization of mononuclear leukocytes (MNLs) in HCC patients treated with Transarterial chemoembolization (TACE). PATIENTS AND METHODS Serum and MNLs were collected from 20 TACE-treated HCC patients before (baseline-control samples) and after treatment with 5 g curcumin capsules , 10 mg piperine and 0.5 mg taurine taken daily for three consecutive months. Immunophenotypic and molecular characterization of MNLs were determined by flow cytometry and quantitative real time PCR, respectively. In addition, serum IFN-γ level was quantified by ELISA. RESULTS After receiving treatment with CPT combination, there was a highly significant increase in IFN- γ levels in the sera of patients when compared to basal line control samples. Additionally, the group receiving combined therapy demonstrated a downregulation in the expression levels of PD-1, in MNLs as compared to controls. MNLs' immunophenotyping revealed a significant decline in CD4+CD25+cells (regulatory T lymphocytes). Furthermore, clinicopathological characteristics revealed a highly significant impact of CPT combination on aspartate aminotransferase (AST), lactate dehydrogenase (LDH) and alpha feto protein (AFP) levels. CONCLUSION This study introduces a promising adjuvant CPT combined treatment as natural agents to enhance the management of HCC patients who are candidates to TACE treatment.
Collapse
Affiliation(s)
| | | | - Heba Effat
- Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, 11796 Cairo, Egypt.
| | - Motawa E. El-Houseini
- Medical Biochemistry and Molecular Biology Unit, Department of Cancer Biology, National Cancer Institute, Cairo University, 11796 Cairo, Egypt.
| | - Randa A. Osman
- Department of Clinical Pathology, National Cancer Institute, Cairo University, 11796 Cairo, Egypt.
| | - Amr Abdelraouf
- Department of Surgery,National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt.
| | - Emad M. Elzayat
- Department of Biotechnology, Faculty of science, Cairo University, 12613 Giza, Egypt.
| |
Collapse
|
136
|
Tang Y, Wei J, Ge X, Yu C, Lu W, Qian Y, Yang H, Fu D, Fang Y, Zhou X, Wang Z, Xiao Q, Ding K. Intratumoral injection of interferon gamma promotes the efficacy of anti-PD1 treatment in colorectal cancer. Cancer Lett 2024; 588:216798. [PMID: 38467181 DOI: 10.1016/j.canlet.2024.216798] [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: 03/20/2023] [Revised: 02/21/2024] [Accepted: 03/05/2024] [Indexed: 03/13/2024]
Abstract
Immune checkpoint inhibitors (ICIs) offer new options for the treatment of patients with solid cancers worldwide. The majority of colorectal cancers (CRC) are proficient in mismatch-repair (pMMR) genes, harboring fewer tumor antigens and are insensitive to ICIs. These tumors are often found to be immune-deserted. We hypothesized that forcing immune cell infiltration into the tumor microenvironment followed by immune ignition by PD1 blockade may initiate a positive immune cycle that can boost antitumor immunity. Bioinformatics using a public database suggested that IFNγ was a key indicator of immune status and prognosis in CRC. Intratumoral administration of IFNγ increased immune cells infiltration into the tumor, but induced PD-L1 expression. A combined treatment strategy using IFNγ and anti-PD-1 antibody significantly increased T cell killing of tumor cells in vitro and showed synergistic inhibition of tumor growth in a mouse model of CRC. CyTOF found drastic changes in the immune microenvironment upon combined immunotherapy. Treatment with IFNγ and anti-PD1 antibody in CT26 tumors significantly increased infiltration of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). IFNγ had a more pronounced effect in decreasing intratumoral M2-like macrophages, while PD1 blockade increased the population of CD8+Ly6C + T cells in the tumor microenvironment, creating a more pro-inflammatory microenvironment. Additionally, PD1 induced increased expression of lymphocyte activating 3 (LAG3) in a significant fraction of CD8+ T cells and Treg cells, indicating potential drug resistance and feedback mechanisms. In conclusion, our work provides preclinical data for the Combined immunotherapy of CRC using intratumoral delivery of IFNγ and systemic anti-PD1 monoclonoal antibody.
Collapse
Affiliation(s)
- Yang Tang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Jingsun Wei
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Chengxuan Yu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Wei Lu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Hang Yang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Yimin Fang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Xinyi Zhou
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Zhanhuai Wang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Zhejiang Province, China), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for CANCER, China; Cancer Center of Zhejiang University, Hangzhou, Zhejiang, 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, China.
| |
Collapse
|
137
|
Petrova L, Bunz F. Interferons in Colorectal Cancer Pathogenesis and Therapy. DISEASES & RESEARCH 2024; 4:31-39. [PMID: 38962090 PMCID: PMC11220628 DOI: 10.54457/dr.202401005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
As key modulators of the immune response, interferons play critical roles following infection and during the pathogenesis of cancer. The idea that these cytokines might be developed as new therapies emerged soon after their discovery. While enthusiasm for this approach to cancer therapy has waxed and waned over the ensuing decades, recent advances in cancer immunotherapy and our improved understanding of the tumor immune environment have led to a resurgence of interest in this unique class of biologic drug. Here, we review how interferons influence the growth of colorectal cancers (CRCs) and highlight new insights into how interferons and drugs that modulate interferon expression might be most effectively deployed in the clinic.
Collapse
Affiliation(s)
- Lucy Petrova
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore Maryland 21287, USA
| | - Fred Bunz
- Department of Radiation Oncology and Molecular Radiation Sciences, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore Maryland 21287, USA
| |
Collapse
|
138
|
Verner JM, Arbuthnott HF, Ramachandran R, Bharadwaj M, Chaudhury N, Jou E. Emerging roles of type 1 innate lymphoid cells in tumour pathogenesis and cancer immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:296-315. [PMID: 38745765 PMCID: PMC11090689 DOI: 10.37349/etat.2024.00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 05/16/2024] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently discovered class of innate immune cells found to have prominent roles in various human immune-related pathologies such as infection and autoimmune diseases. However, their role in cancer was largely unclear until recently, where several emerging studies over the past few years unanimously demonstrate ILCs to be critical players in tumour immunity. Being the innate counterpart of T cells, ILCs are potent cytokine producers through which they orchestrate the overall immune response upstream of adaptive immunity thereby modulating T cell function. Out of the major ILC subsets, ILC1s have gained significant traction as potential immunotherapeutic candidates due to their central involvement with the anti-tumour type 1 immune response. ILC1s are potent producers of the well-established anti-tumour cytokine interferon γ (IFNγ), and exert direct cytotoxicity against cancer cells in response to the cytokine interleukin-15 (IL-15). However, in advanced diseases, ILC1s are found to demonstrate an exhausted phenotype in the tumour microenvironment (TME) with impaired effector functions, characterised by decreased responsiveness to cytokines and reduced IFNγ production. Tumour cells produce immunomodulatory cytokines such as transforming growth factor β (TGFβ) and IL-23, and through these suppress ILC1 anti-tumour actfivities and converts ILC1s to pro-tumoural ILC3s respectively, resulting in disease progression. This review provides a comprehensive overview of ILC1s in tumour immunity, and discusses the exciting prospects of harnessing ILC1s for cancer immunotherapy, either alone or in combination with cytokine-based treatment. The exciting prospects of targeting the upstream innate immune system through ILC1s may surmount the limitations associated with adaptive immune T cell-based strategies used in the clinic currently, and overcome cancer immunotherapeutic resistance.
Collapse
Affiliation(s)
| | | | - Raghavskandhan Ramachandran
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom
- Balliol College, University of Oxford, OX1 3BJ Oxford, United Kingdom
| | - Manini Bharadwaj
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Natasha Chaudhury
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Eric Jou
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom
- Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
- Kellogg College, University of Oxford, OX2 6PN Oxford, United Kingdom
| |
Collapse
|
139
|
Verner JM, Arbuthnott HF, Ramachandran R, Bharadwaj M, Chaudhury N, Jou E. Emerging roles of type 1 innate lymphoid cells in tumour pathogenesis and cancer immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:296-315. [DOI: 10.37349/etat.2023.00219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/14/2023] [Indexed: 01/04/2025] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently discovered class of innate immune cells found to have prominent roles in various human immune-related pathologies such as infection and autoimmune diseases. However, their role in cancer was largely unclear until recently, where several emerging studies over the past few years unanimously demonstrate ILCs to be critical players in tumour immunity. Being the innate counterpart of T cells, ILCs are potent cytokine producers through which they orchestrate the overall immune response upstream of adaptive immunity thereby modulating T cell function. Out of the major ILC subsets, ILC1s have gained significant traction as potential immunotherapeutic candidates due to their central involvement with the anti-tumour type 1 immune response. ILC1s are potent producers of the well-established anti-tumour cytokine interferon γ (IFNγ), and exert direct cytotoxicity against cancer cells in response to the cytokine interleukin-15 (IL-15). However, in advanced diseases, ILC1s are found to demonstrate an exhausted phenotype in the tumour microenvironment (TME) with impaired effector functions, characterised by decreased responsiveness to cytokines and reduced IFNγ production. Tumour cells produce immunomodulatory cytokines such as transforming growth factor β (TGFβ) and IL-23, and through these suppress ILC1 anti-tumour actfivities and converts ILC1s to pro-tumoural ILC3s respectively, resulting in disease progression. This review provides a comprehensive overview of ILC1s in tumour immunity, and discusses the exciting prospects of harnessing ILC1s for cancer immunotherapy, either alone or in combination with cytokine-based treatment. The exciting prospects of targeting the upstream innate immune system through ILC1s may surmount the limitations associated with adaptive immune T cell-based strategies used in the clinic currently, and overcome cancer immunotherapeutic resistance.
Collapse
Affiliation(s)
| | | | - Raghavskandhan Ramachandran
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom; Balliol College, University of Oxford, OX1 3BJ Oxford, United Kingdom
| | - Manini Bharadwaj
- exham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Natasha Chaudhury
- exham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom
| | - Eric Jou
- Medical Sciences Division, Oxford University Hospitals, OX3 9DU Oxford, United Kingdom; Wexham Park Hospital, Frimley Health NHS Foundation Trust, SL2 4HL Slough, United Kingdom; Kellogg College, University of Oxford, OX2 6PN Oxford, United Kingdom
| |
Collapse
|
140
|
Chae SY, Shin H, Woo J, Kang S, Lee SM, Min DH. Metabolic Modulation of Kynurenine Based on Kynureninase-Loaded Nanoparticle Depot Overcomes Tumor Immune Evasion in Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:18490-18502. [PMID: 38573937 DOI: 10.1021/acsami.4c00513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Evading recognition of immune cells is a well-known strategy of tumors used for their survival. One of the immune evasion mechanisms is the synthesis of kynurenine (KYN), a metabolite of tryptophan, which suppresses the effector T cells. Therefore, lowering the KYN concentration can be an efficient antitumor therapy by restoring the activity of immune cells. Recently, kynureninase (KYNase), which is an enzyme transforming KYN into anthranilate, was demonstrated to show the potential to decrease KYN concentration and inhibit tumor growth. However, due to the limited bioavailability and instability of proteins in vivo, it has been challenging to maintain the KYNase concentration sufficiently high in the tumor microenvironment (TME). Here, we developed a nanoparticle system loaded with KYNase, which formed a Biodegradable and Implantable Nanoparticle Depot named 'BIND' following subcutaneous injection. The BIND sustainably supplied KYNase around the TME while located around the tumor, until it eventually degraded and disappeared. As a result, the BIND system enhanced the proliferation and cytokine production of effector T cells in the TME, followed by tumor growth inhibition and increased mean survival. Finally, we showed that the BIND carrying KYNase significantly synergized with PD-1 blockade in three mouse models of colon cancer, breast cancer, and melanoma.
Collapse
Affiliation(s)
- Se-Youl Chae
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Hojeong Shin
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiwon Woo
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Seounghun Kang
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Soo Min Lee
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| | - Dal-Hee Min
- Department of Chemistry, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
141
|
Tang M, Jiang S, Huang X, Ji C, Gu Y, Qi Y, Xiang Y, Yao E, Zhang N, Berman E, Yu D, Qu Y, Liu L, Berry D, Yao Y. Integration of 3D bioprinting and multi-algorithm machine learning identified glioma susceptibilities and microenvironment characteristics. Cell Discov 2024; 10:39. [PMID: 38594259 PMCID: PMC11003988 DOI: 10.1038/s41421-024-00650-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/18/2024] [Indexed: 04/11/2024] Open
Abstract
Glioma, with its heterogeneous microenvironments and genetic subtypes, presents substantial challenges for treatment prediction and development. We integrated 3D bioprinting and multi-algorithm machine learning as a novel approach to enhance the assessment and understanding of glioma treatment responses and microenvironment characteristics. The bioprinted patient-derived glioma tissues successfully recapitulated molecular properties and drug responses of native tumors. We then developed GlioML, a machine learning workflow incorporating nine distinct algorithms and a weighted ensemble model that generated robust gene expression-based predictors, each reflecting the diverse action mechanisms of various compounds and drugs. The ensemble model superseded the performance of all individual algorithms across diverse in vitro systems, including sphere cultures, complex 3D bioprinted multicellular models, and 3D patient-derived tissues. By integrating bioprinting, the evaluative scope of the treatment expanded to T cell-related therapy and anti-angiogenesis targeted therapy. We identified promising compounds and drugs for glioma treatment and revealed distinct immunosuppressive or angiogenic myeloid-infiltrated tumor microenvironments. These insights pave the way for enhanced therapeutic development for glioma and potentially for other cancers, highlighting the broad application potential of this integrative and translational approach.
Collapse
Affiliation(s)
- Min Tang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA.
| | - Shan Jiang
- Department of Statistics, University of California Davis, Davis, CA, USA
| | - Xiaoming Huang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Chunxia Ji
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yexin Gu
- Cyberiad Biotechnology Ltd., Shanghai, China
| | - Ying Qi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yi Xiang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Emmie Yao
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
| | - Nancy Zhang
- Department of Human Biology, University of California San Diego, La Jolla, CA, USA
| | - Emma Berman
- Department of Bioengineering, University of California Berkeley, Berkeley, CA, USA
| | - Di Yu
- Department of Human Biology, University of California San Diego, La Jolla, CA, USA
| | - Yunjia Qu
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Longwei Liu
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - David Berry
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, USA
- Department of Orthopaedic Surgery, University of California San Diego, La Jolla, CA, USA
| | - Yu Yao
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Immunology Laboratory, Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
142
|
Visallini TS, Parameswari RP. Retrospective Case Study on the Evaluation of Inflammatory Markers, Macrophage Inhibitory Protein-1α and Interferon-γ in Sleep Deprivation Condition. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1326-S1329. [PMID: 38882737 PMCID: PMC11174169 DOI: 10.4103/jpbs.jpbs_607_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 06/18/2024] Open
Abstract
Background and Aim Sleep is an important physiological process that is necessary for the normal functioning of the body. Sleep greatly affects all aspects of our body, including the immune pathways or immune response system of our body, which plays a determinant role in the development and progression of chronic inflammatory diseases. In this study, we worked to find the relation between sleep deprivation and levels of pro-inflammatory markers macrophage inflammatory protein 1-alpha (MIP-1α) and interferon gamma (IFN-γ). To find the relation between sleep deprivation and levels of pro-inflammatory markers MIP-1α and IFN-γ. Objective To find the relation between sleep deprivation and levels of pro-inflammatory markers MIP-1α and IFN-γ. Materials and Methods The study was conducted with 40 individuals as participants, of which 20 were sleep-deprived (SD), and 20 had adequate amounts of sleep. The sleep duration details of the individuals were obtained by questionnaire. Blood was withdrawn from all the subjects after due consent from them. Plasma was separated and was used to evaluate their MIP-1α levels and IFN-γ levels. Results The MIP-1α levels and levels of IFN-γ were found to be significantly elevated in the SD individuals than that of individuals who had adequate sleep. Conclusion Sleep loss and sleep deprivation are associated with altered expressions of key regulatory factors and upregulation of pro-inflammatory cytokines production. Thus, sleep deprivation can be considered to be one of the major contributors to the development and progression of various chronic inflammatory diseases.
Collapse
Affiliation(s)
- T S Visallini
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Thandalam, Chennai, Tamil Nadu, India
| | - R P Parameswari
- Department of Pharmacology, Centre for Transdisciplinary Research, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
| |
Collapse
|
143
|
Das D, Banerjee A, Manna K, Sarkar D, Shil A, Sikdar Ne E Bhakta M, Mukherjee S, Maji BK. Quercetin counteracts monosodium glutamate to mitigate immunosuppression in the thymus and spleen via redox-guided cellular signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:155226. [PMID: 38387276 DOI: 10.1016/j.phymed.2023.155226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/13/2023] [Accepted: 11/18/2023] [Indexed: 02/24/2024]
Abstract
BACKGROUND Chronic inflammation brought on by oxidative stress can result in several immunopathologies. Natural compounds with antioxidant characteristics, like quercetin, have shown effectiveness in reducing oxidative damage and regulating the immune response. PURPOSE The commonly used food additive monosodium glutamate (M) causes immunosuppression by disrupting redox equilibrium and inducing oxidative stress. The goal of this work is to examine the therapeutic potential of quercetin against immunotoxicity brought on by M, revealing the molecular route implicated in such immunopathology by targeting the thymus and spleen, to support the development of future anti-inflammatory and antioxidant therapies. STUDY DESIGN AND METHODS M-fed rats were employed as an immunotoxicity model and were supplemented with quercetin for four weeks. Hematological and biochemical parameters were measured; H&E staining, immunohistochemistry, flow cytometry, real-time quantitative PCR, and western blotting were performed. RESULTS Based on the findings, TLR4 was activated by M to cause oxidative stress-mediated inflammation, which was alleviated by the supplementation of quercetin by modulating redox homeostasis to neutralize free radicals and suppress the inflammatory response. To prevent M-induced inflammation, quercetin demonstrated anti-inflammatory functions by blocking NF-kB activation, lowering the production of pro-inflammatory cytokines, and increasing the release of anti-inflammatory cytokines. By normalizing lipid profiles and lowering the potential risk of immunological deficiency caused by M, quercetin also improves lipid metabolism. Additionally, it has shown potential for modifying insulin levels, suggesting a possible function in controlling M-induced alteration in glucose metabolism. The addition of quercetin to M enhanced the immune response by improving immunoglobulin levels and CD4/CD8 expression in the thymus and spleen. Additionally, quercetin inhibited apoptosis by controlling mitochondrial caspase-mediated cellular signaling, suggesting that it may be able to halt cell death in M-fed rats. CONCLUSION The results of this study first indicate that quercetin, via modulating redox-guided cellular signaling, has a promising role in reducing immune disturbances. This study illuminates the potential of quercetin as a safe, natural remedy for immunopathology caused by M, including thymic hypoplasia and/or splenomegaly, and paves the way for future anti-inflammatory and antioxidant supplements.
Collapse
Affiliation(s)
- Debasmita Das
- Department of Physiology (UG & PG), Serampore College, 9 William Carey Road, Serampore, Hooghly-712201, West Bengal, India
| | - Arnab Banerjee
- Department of Physiology (UG & PG), Serampore College, 9 William Carey Road, Serampore, Hooghly-712201, West Bengal, India
| | - Krishnendu Manna
- Department of Food & Nutrition, University of Kalyani, Kalyani, Nadia, West Bengal, India
| | - Deotima Sarkar
- Department of Bacteriology, National Institute of Cholera and Enteric Diseases, Indian Council of Medical Research (ICMR-NICED), Kolkata 700010, India
| | - Aparna Shil
- Microbiology, Nutrition and Dietetics Laboratory, Physiology Unit, Department of Life Sciences, Presidency University, Kolkata-700073, India
| | - Mausumi Sikdar Ne E Bhakta
- Microbiology, Nutrition and Dietetics Laboratory, Physiology Unit, Department of Life Sciences, Presidency University, Kolkata-700073, India
| | - Sandip Mukherjee
- Department of Physiology (UG & PG), Serampore College, 9 William Carey Road, Serampore, Hooghly-712201, West Bengal, India
| | - Bithin Kumar Maji
- Department of Physiology (UG & PG), Serampore College, 9 William Carey Road, Serampore, Hooghly-712201, West Bengal, India.
| |
Collapse
|
144
|
Iyer M, Ravichandran N, Karuppusamy PA, Gnanarajan R, Yadav MK, Narayanasamy A, Vellingiri B. Molecular insights and promise of oncolytic virus based immunotherapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:419-492. [PMID: 38762277 DOI: 10.1016/bs.apcsb.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Discovering a therapeutic that can counteract the aggressiveness of this disease's mechanism is crucial for improving survival rates for cancer patients and for better understanding the most different types of cancer. In recent years, using these viruses as an anticancer therapy has been thought to be successful. They mostly work by directly destroying cancer cells, activating the immune system to fight cancer, and expressing exogenous effector genes. For the treatment of tumors, oncolytic viruses (OVs), which can be modified to reproduce only in tumor tissues and lyse them while preserving the healthy non-neoplastic host cells and reinstating antitumor immunity which present a novel immunotherapeutic strategy. OVs can exist naturally or be created in a lab by altering existing viruses. These changes heralded the beginning of a new era of less harmful virus-based cancer therapy. We discuss three different types of oncolytic viruses that have already received regulatory approval to treat cancer as well as clinical research using oncolytic adenoviruses. The primary therapeutic applications, mechanism of action of oncolytic virus updates, future views of this therapy will be covered in this chapter.
Collapse
Affiliation(s)
- Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Roselin Gnanarajan
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
145
|
Yin KL, Chu KJ, Li M, Duan YX, Yu YX, Kang MQ, Fu D, Liao R. Immune Regulatory Networks and Therapy of γδ T Cells in Liver Cancer: Recent Trends and Advancements. J Clin Transl Hepatol 2024; 12:287-297. [PMID: 38426194 PMCID: PMC10899867 DOI: 10.14218/jcth.2023.00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 03/02/2024] Open
Abstract
The roles of γδ T cells in liver cancer, especially in the potential function of immunotherapy due to their direct cytotoxic effects on tumor cells and secretion of important cytokines and chemokines, have aroused research interest. This review briefly describes the basic characteristics of γδ T cells, focusing on their diverse effects on liver cancer. In particular, different subtypes of γδ T cells have diverse or even opposite effects on liver cancer. We provide a detailed description of the immune regulatory network of γδ T cells in liver cancer from two aspects: immune components and nonimmune components. The interactions between various components in this immune regulatory network are dynamic and pluralistic, ultimately determining the biological effects of γδ T cells in liver cancer. We also integrate the current knowledge of γδ T-cell immunotherapy for liver cancer treatment, emphasizing the potential of these cells in liver cancer immunotherapy.
Collapse
Affiliation(s)
- Kun-Li Yin
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai-Jian Chu
- Biliary Surgical Department I, the Eastern Hepatobiliary Surgical Hospital, Naval Medical University, Shanghai, China
| | - Ming Li
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu-Xin Duan
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan-Xi Yu
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mei-Qing Kang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Da Fu
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Rui Liao
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
146
|
Trimaglio G, Sneperger T, Raymond BBA, Gilles N, Näser E, Locard-Paulet M, Ijsselsteijn ME, Brouwer TP, Ecalard R, Roelands J, Matsumoto N, Colom A, Habch M, de Miranda NFCC, Vergnolle N, Devaud C, Neyrolles O, Rombouts Y. The C-type lectin DCIR contributes to the immune response and pathogenesis of colorectal cancer. Sci Rep 2024; 14:7199. [PMID: 38532110 DOI: 10.1038/s41598-024-57941-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 03/22/2024] [Indexed: 03/28/2024] Open
Abstract
Development and progression of malignancies are accompanied and influenced by alterations in the surrounding immune microenvironment. Understanding the cellular and molecular interactions between immune cells and cancer cells has not only provided important fundamental insights into the disease, but has also led to the development of new immunotherapies. The C-type lectin Dendritic Cell ImmunoReceptor (DCIR) is primarily expressed by myeloid cells and is an important regulator of immune homeostasis, as demonstrated in various autoimmune, infectious and inflammatory contexts. Yet, the impact of DCIR on cancer development remains largely unknown. Analysis of available transcriptomic data of colorectal cancer (CRC) patients revealed that high DCIR gene expression is associated with improved patients' survival, immunologically "hot" tumors and high immunologic constant of rejection, thus arguing for a protective and immunoregulatory role of DCIR in CRC. In line with these correlative data, we found that deficiency of DCIR1, the murine homologue of human DCIR, leads to the development of significantly larger tumors in an orthotopic murine model of CRC. This phenotype is accompanied by an altered phenotype of tumor-associated macrophages (TAMs) and a reduction in the percentage of activated effector CD4+ and CD8+ T cells in CRC tumors of DCIR1-deficient mice. Overall, our results show that DCIR promotes antitumor immunity in CRC, making it an attractive target for the future development of immunotherapies to fight the second deadliest cancer in the world.
Collapse
Affiliation(s)
- Giulia Trimaglio
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Tamara Sneperger
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Benjamin B A Raymond
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Nelly Gilles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Emmanuelle Näser
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Marie Locard-Paulet
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Thomas P Brouwer
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Romain Ecalard
- INSERM US006 ANEXPLO/CREFRE, Purpan Hospital, Toulouse, France
| | - Jessica Roelands
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Naoki Matsumoto
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - André Colom
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Myriam Habch
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | | | - Nathalie Vergnolle
- Institut de Recherche en Santé Digestive, IRSD, Université de Toulouse, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Christel Devaud
- Institut de Recherche en Santé Digestive, IRSD, Université de Toulouse, INSERM, INRAe, ENVT, UPS, Toulouse, France
| | - Olivier Neyrolles
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Yoann Rombouts
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| |
Collapse
|
147
|
Xue Y, Yan X, Li D, Dong S, Ping Y. Proinflammatory polarization of engineered heat-inducible macrophages reprogram the tumor immune microenvironment during cancer immunotherapy. Nat Commun 2024; 15:2270. [PMID: 38491004 PMCID: PMC10943244 DOI: 10.1038/s41467-024-46210-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
The success of macrophage-based adoptive cell therapy is largely constrained by poor polarization from alternatively activated (M2-like) to classically activated (M1-like) phenotype in the immunosuppressive tumor microenvironment (TME). Here, we show that the engineered macrophage (eMac) with a heat-inducible genetic switch can induce both self-polarization of adoptively transferred eMac and re-polarization of tumour-associated macrophages in response to mild temperature elevation in a mouse model. The locoregional production of proinflammatory cytokines by eMac in the TME dose not only induces the strong polarization of macrophages into a classically activated phenotype, but also ensures that the side effects typical for systemically administrate proinflammatory cytokines are avoided. We also present a wearable warming device which is adaptable for human patients and can be remotely controlled by a smartphone. In summary, our work represents a safe and efficient adoptive transfer immunotherapy method with potential for human translation.
Collapse
Affiliation(s)
- Yanan Xue
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaojie Yan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China
| | - Da Li
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Shurong Dong
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuan Ping
- Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University, Hangzhou, 311121, China.
| |
Collapse
|
148
|
Bhatt DK, Meuleman SL, Hoogeboom BN, Daemen T. Oncolytic alphavirus replicons mediated recruitment and activation of T cells. iScience 2024; 27:109253. [PMID: 38425844 PMCID: PMC10904282 DOI: 10.1016/j.isci.2024.109253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
Oncolytic viruses show promise in enhancing tumor immunogenicity by releasing immunogenic signals during tumor cell infection and lysis. In this study, we improved the virus-induced tumor immunogenicity of recombinant Semliki Forest virus (rSFV)-based replicon particles by encoding immunogenic cytokines such as C-X-C motif chemokine ligand 10 (CXCL10), FMS-like tyrosine kinase 3 ligand (Flt3L), or interferon-gamma (IFN-ƴ). Real-time imaging and flow cytometry of human cancer cell-based monolayer and spheroid cultures, using LNCaP or PANC-1 cells, revealed effective infection and transgene expression in both models. LNCaP cells exhibited higher and earlier rSFV infection compared to PANC-1 cells. While infected LNCaP cells effectively triggered immune recruitment and T cell activation even without encoding cytokines, PANC-1 cells demonstrated improved immune responses only when infected with replicons encoding cytokines, particularly IFN-ƴ, which enhanced tumor immunogenicity irrespective of cancer cell susceptibility to infection. Our study demonstrates that despite innate phenotypic disparities in cancer cells, rSFV-based replicons encoding cytokines can potentially generate effective immune responses in the tumor.
Collapse
Affiliation(s)
- Darshak K. Bhatt
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Saskia L. Meuleman
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, the Netherlands
| |
Collapse
|
149
|
Zhao B, Zheng X, Wang Y, Cheng N, Zhong Y, Zhou Y, Huang J, Wang F, Qi X, Zhuang Q, Wang Y, Liu X. Lnc-CCNH-8 promotes immune escape by up-regulating PD-L1 in hepatocellular carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102125. [PMID: 38356866 PMCID: PMC10865404 DOI: 10.1016/j.omtn.2024.102125] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy with poor prognosis. In recent years, immune checkpoint inhibitors (ICIs) have enabled breakthroughs in the clinical treatment of patients with HCC, but the overall response rate to ICIs in HCC patients is still low, and no validated biomarker is available to guide clinical decision making. Here, we demonstrated that the long non-coding RNA Lnc-CCNH-8 is highly expressed in HCC and correlates with poor prognosis. Functionally, elevated Lnc-CCNH-8 inactivated co-cultured T cells in vitro and compromised antitumor immunity in an immunocompetent mouse model. Mechanistically, up-regulated Lnc-CCNH-8 can sponge microRNA (miR)-217 to regulate the expression of PD-L1. In addition, Lnc-CCNH-8 can also stabilize PD-L1 through miR-3173/PKP3 axis. Furthermore, mice bearing tumors with high Lnc-CCNH-8 expression had significant therapeutic sensitivity to anti-PD-L1 monoclonal antibody treatment. More important, HCC patients with high levels of plasma exosomal Lnc-CCNH-8 had a better therapeutic response to ICIs. Taken together, our results reveal the function of Lnc-CCNH-8 in inducing immune escape from CD8+ T-cell-mediated killing by up-regulating PD-L1 in a miR-217/miR-3173-dependent manner, which also reveals a novel mechanism of PD-L1 regulation in HCC, and exosomal Lnc-CCNH-8 can serve as a predictive marker for immunotherapy response in HCC.
Collapse
Affiliation(s)
- Bixing Zhao
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xiaoyuan Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yang Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Niangmei Cheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yue Zhong
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
| | - Yang Zhou
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Jingyun Huang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Fei Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xin Qi
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
| | - Qiuyu Zhuang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Yingchao Wang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P.R. China
- Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P.R. China
- Fujian Provincial Clinical Research Center for Hepatobiliary and Pancreatic Tumors, Fuzhou 350025, P.R. China
| |
Collapse
|
150
|
Luo R, Wan Y, Liu G, Chen J, Luo X, Li Z, Su D, Lu N, Luo Z. Engineering Self-Assembling Peptide Hydrogel to Enhance the Capacity of Dendritic Cells to Activate In Vivo T-Cell Immunity. Biomacromolecules 2024; 25:1408-1428. [PMID: 38236703 DOI: 10.1021/acs.biomac.3c00511] [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: 03/12/2024]
Abstract
The efficacy of the dendritic cell (DC) has failed to meet expectations thus far, and crucial problems such as the immature state of DCs, low targeting efficiency, insufficient number of dendritic cells, and microenvironment are still the current focus. To address these problems, we developed two self-assembling peptides, RLDI and RQDT, that mimic extracellular matrix (ECM). These peptides can be self-assembled into highly ordered three-dimensional nanofiber scaffold structures, where RLDI can form gelation immediately. In addition, we found that RLDI and RQDT enhance the biological function of DCs, including releasing antigens sustainably, adhering to DCs, promoting the maturation of DCs, and increasing the ability of DC antigen presentation. Moreover, peptide hydrogel-based DC treatment significantly achieved prophylactic and treatment effects on colon cancer. These results have certain implications for the design of new broad-spectrum vaccines in the future.
Collapse
Affiliation(s)
- Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa 52242, United States
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhaoxu Li
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|