1
|
Dakal TC, Philip RR, Bhushan R, Sonar PV, Rajagopal S, Kumar A. Genetic and epigenetic regulation of non-coding RNAs: Implications in cancer metastasis, stemness and drug resistance. Pathol Res Pract 2024; 266:155728. [PMID: 39657397 DOI: 10.1016/j.prp.2024.155728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/11/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024]
Abstract
Cancer stem cells (CSCs) have a crucial function in the initiation, advancement, and resistance to therapy of tumors. Recent findings indicate that non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), play a complex role in controlling the features of cancer stem cells (CSCs). Non-coding RNAs (ncRNAs) play a crucial role in controlling important characteristics of stem cells, such as their ability to renew themselves, differentiate into distinct cell types, and resist therapy. This article provides an overview of the current understanding of the complex relationship between non-coding RNAs (ncRNAs), namely microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), and cancer stem cells (CSCs). Particular microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are involved in regulating important signaling pathways like as Wnt, Notch, and Hedgehog, which control stem cell-like characteristics. The miR-34, miR-200, and let-7 families specifically aim at inhibiting the process of self-renewal and epithelial-to-mesenchymal transition. On the other hand, long non-coding RNAs (lncRNAs) such as H19, HOTAIR, and MALAT1 play a role in modifying the epigenetic landscape, hence enhancing the characteristics of stemness. This article also offers a thorough examination of the role of non-coding RNAs (ncRNAs) in regulating cancer stemness, emphasizing their impact on crucial biochemical pathways, epigenetic changes, and therapeutic implications. Comprehending the interaction between non-coding RNAs (ncRNAs) and cancer stem cells (CSCs) provides fresh perspectives on possible focused treatments for fighting aggressive and resistant malignancies. Gaining a comprehensive understanding of the connection between non-coding RNA (ncRNA) and cancer stem cells (CSC) offers valuable insights for the development of novel and precise treatments to combat aggressive cancers that are resistant to conventional therapies. In addition, the combination of ncRNA therapies with conventional methods like as chemotherapy or epigenetic medicines could result in synergistic effects. Nevertheless, there are still obstacles to overcome in terms of delivery, effectiveness, and safety. In summary, the interaction between non-coding RNA and cancer stemness shows potential as a targeted treatment approach in the field of precision oncology. This calls for additional investigation and use in clinical settings.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India.
| | - Reya Rene Philip
- Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Chennai, India
| | - Ravi Bhushan
- Department of Zoology, M.S. College, Motihari, Bihar 845401, India
| | | | - Senthilkumar Rajagopal
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru, Karnataka, India
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India; Manipal Academy of Higher Education (MAHE), Manipal, Karnataka 576104, India.
| |
Collapse
|
2
|
Christakoudi S, Tsilidis KK, Gunter MJ, Riboli E. Allometric fat mass index and alanine aminotransferase attenuate the associations of platelet parameters with lung cancer risk. Sci Rep 2024; 14:26318. [PMID: 39487349 PMCID: PMC11530616 DOI: 10.1038/s41598-024-78281-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024] Open
Abstract
We have previously shown that body mass index attenuates a positive association of platelet count (PLT) and inverse of mean platelet volume (MPV) with lung cancer risk in men. It is unclear whether fat mass, lean mass, or liver function tests (LFTs) show similar attenuations. Using bioelectrical impedance measurements (UK Biobank cohort) and multivariable Cox proportional hazards models, we examined the associations of allometric fat-mass index (AFI, fat mass adjusted for height), allometric lean-mass index (ALI, fat-free mass adjusted for height and fat mass), and LFTs with lung cancer risk and their multiplicative and additive interactions with platelet parameters. Based on 1573 lung cancer cases in men and 1473 in women with body composition measurements (1541 in men; 1428 in women with biomarker measurements), AFI in women, ALI in both sexes, alanine aminotransferase (ALT) and total bilirubin in men were inversely associated, while gamma-glutamyl transferase in men and alkaline phosphatase in both sexes were positively associated with lung cancer risk. Only AFI and ALT interacted inversely with PLT and positively with MPV in men. The attenuation of the associations of platelet parameters with lung cancer risk by high-AFI and high-ALT in men suggests that adiposity-related factors hinder lung-cancer-related platelet associations.
Collapse
Affiliation(s)
- Sofia Christakoudi
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK.
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Marc J Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, White City Campus, 90 Wood Lane, London, W12 0BZ, UK
| |
Collapse
|
3
|
Xu H, Long J, Qi X, Li P, Yan C, Wang L, Jin Y, Liu H. Galectin-9 activates host immune response and improve immunoprotection of Onychostoma macrolepis against Aeromonas hydrophila infection. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109929. [PMID: 39341377 DOI: 10.1016/j.fsi.2024.109929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/05/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
Galectin-9 (Gal-9) belongs to a family of the glycan-binding proteins (GBPs) and is known to restrict bacterial activity via interacting with pathogen associated molecular pattern (PAMPs). However, the underlying immune mechanism of endogenous Gal-9 on fish against bacterial infection is still unclear. In this study, effects of Gal-9 from Onychostoma macrolepis (OmGal-9) on expression of immune-related genes were measured by HEK293T. The immune response of O. macrolepis with OmGal-9 overexpression to Aeromonas hydrophila (A. hydrophila) infection (1.65 × 108 CFU/mL) was evaluated by tissue bacterial load, fish survival rate and transcriptome analysis. The results showed that OmGal-9 displayed a punctate distribution in the nucleus and cytoplasm of HEK293T cells. Compared to cells transfected with the empty vector (EV group), recombinant plasmid pEGFP-Gal9 treatment (Gal9 group) significantly down-regulated the expression of immune-related genes TNFα, STAT3, MyD88, LCK, and p52 of HEK293T cells stimulated with LPS at 24 h, while up-regulated IκBα and caspase-1 (P < 0.05). The activities of catalase (CAT), superoxide dismutase (SOD), the total antioxidant capacity (T-AOC), alkaline phosphatase (AKP), acid phosphatase (ACP), and lysozyme (LZM) of O. macrolepis were significantly increased on 7 days in Gal9 group compared to EV group (P < 0.05). The bacterial load of liver, spleen, and kidney of O. macrolepis infected with A. hydrophila in Gal9 group at 24 h was significantly lower than that in EV group (P < 0.05), and the survival rate had increased from 15 % to 35 %. A comparative transcriptome analysis between the Gal9 and EV group identified 305 differentially expressed genes (DEGs). The analysis showed that OmGal-9 might play an important regulatory role in glycolysis/gluconeogenesis, fatty acid degradation, and ascorbate and aldarate metabolism. Moreover, the immune-related DEGs were predominantly enriched in eleven pathways, with the most important three of them being linked to innate immunity: NOD-like, C-type lectin and Toll-like receptor signaling pathway. Taking together, OmGal-9 can enhance the resistance of fish to bacterial diseases by improving immune system function and activating immune-related pathways.
Collapse
Affiliation(s)
- Hongzhou Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Jingfei Long
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaoyu Qi
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ping Li
- Power China Northwest Engineering Corporation Limited, Xi'an, 710065, Shaanxi, China
| | - Chenyang Yan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Lixin Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yuanjiang Jin
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Haixia Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
4
|
Tran B, Voskoboynik M, Bendell J, Gutierrez M, Lemech C, Day D, Frentzas S, Garrido-Laguna I, Standifer N, Wang F, Ferte C, Wang Y, Das M, Carneiro BA. A phase 1 study of the CD40 agonist MEDI5083 in combination with durvalumab in patients with advanced solid tumors. Immunotherapy 2024; 16:759-774. [PMID: 39264730 PMCID: PMC11421296 DOI: 10.1080/1750743x.2024.2359359] [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: 12/12/2023] [Accepted: 05/21/2024] [Indexed: 09/14/2024] Open
Abstract
Aim: This first-in-human study evaluated safety and efficacy of CD40 agonist MEDI5083 with durvalumab in patients with advanced solid tumors.Methods: Patients received MEDI5083 (3-7.5 mg subcutaneously every 2 weeks × 4 doses) and durvalumab (1500 mg every 4 weeks) either sequentially (N = 29) or concurrently (N = 9). Primary end point was safety; secondary end points included efficacy.Results: Thirty-eight patients received treatment. Most common adverse events (AEs) were injection-site reaction (ISR; sequential: 86%; concurrent: 100%), fatigue (41%; 33%), nausea (20.7%; 55.6%) and decreased appetite (24.1%; 33.3%). Nine patients had MEDI5083-related grade ≥3 AEs with ISR being the most common. Two patients experienced dose limiting toxicities (ISR). One death occurred due to a MEDI5083-related AE. MEDI5083 maximum tolerated dose was 5 mg. Objective response rate was 2.8% (1 partial response and 11 stable disease).Conclusion: MEDI5083 toxicity profile limits its further development.
Collapse
Affiliation(s)
- Ben Tran
- Peter MacCallum Cancer Centre, Melbourne, 8006, Australia
| | - Mark Voskoboynik
- Nucleus Network, Melbourne, 3004, Australia
- Monash University, Melbourne, 3004, Australia
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN 37203, USA
| | | | - Charlotte Lemech
- Scientia Clinical Research, Randwick, 2031, Australia
- The University of New South Wales, Sydney, 2052, Australia
| | - Daphne Day
- Monash University, Melbourne, 3004, Australia
- Monash Medical Centre, Clayton, 3800, Australia
| | - Sophia Frentzas
- Monash University, Melbourne, 3004, Australia
- Monash Medical Centre, Clayton, 3800, Australia
| | | | - Nathan Standifer
- Integrated Bioanalysis, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, South San Francisco, CA 94080, USA
| | - Fujun Wang
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD 20878,USA
| | | | - Yue Wang
- AstraZeneca, Gaithersburg, MD 20878,USA
| | | | - Benedito A Carneiro
- Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, RI 02903,USA
| |
Collapse
|
5
|
Wang M, Zhu Z, Wu X, Cheong K, Li X, Yu W, Yao Y, Wu J, Cao Z. Bioactive Polysaccharides from Gracilaria lemaneiformis: Preparation, Structures, and Therapeutic Insights. Foods 2024; 13:2782. [PMID: 39272547 PMCID: PMC11395005 DOI: 10.3390/foods13172782] [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: 07/31/2024] [Revised: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Gracilaria lamaneiformis, a red seaweed, is an abundant source of bioactive polysaccharides with significant health-promoting properties. Nevertheless, the broad application of G. lamaneiformis in the nutraceutical and pharmaceutical sectors remains constrained due to the absence of comprehensive data. This review provides a detailed examination of the preparation methods, structural characteristics, and biological activities of G. lamaneiformis polysaccharides (GLPs). We explore both conventional and advanced extraction techniques, highlighting the efficiency and yield improvements achieved through methods such as microwave-, ultrasonic-, and enzyme-assisted extraction. The structural elucidation of GLPs using modern analytical techniques, including high-performance liquid chromatography, gas chromatography, and nuclear magnetic resonance spectroscopy, is discussed, providing comprehensive insights into their molecular composition and configuration. Furthermore, we critically evaluate the diverse biological activities of GLPs, including their antioxidant, anti-inflammatory, antitumor, and gut microbiota modulation properties. This review underscores the therapeutic potential of GLPs and suggests future research directions to fully harness their health benefits.
Collapse
Affiliation(s)
- Min Wang
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhen Zhu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaocheng Wu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Kitleong Cheong
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Xiaohua Li
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Wanli Yu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Yinlin Yao
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Jiang Wu
- College of Coastal Agriculture Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhanhui Cao
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
6
|
Ribeiro JM, Mendes J, Gante I, Figueiredo-Dias M, Almeida V, Gomes A, Regateiro FJ, Regateiro FS, Caramelo F, Silva HC. Two Different Immune Profiles Are Identified in Sentinel Lymph Nodes of Early-Stage Breast Cancer. Cancers (Basel) 2024; 16:2881. [PMID: 39199652 PMCID: PMC11352239 DOI: 10.3390/cancers16162881] [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: 07/18/2024] [Revised: 08/11/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
In the management of early-stage breast cancer (BC), lymph nodes (LNs) are typically characterised using the One-Step Nucleic Acid Amplification (OSNA) assay, a standard procedure for assessing subclinical metastasis in sentinel LNs (SLNs). The pivotal role of LNs in coordinating the immune response against BC is often overlooked. Our aim was to improve prognostic information provided by the OSNA assay and explore immune-related gene signatures in SLNs. The expression of an immune gene panel was analysed in SLNs from 32 patients with Luminal A early-stage BC (cT1-T2 N0). Using an unsupervised approach based on these expression values, this study identified two clusters, regardless of the SLN invasion: one evidencing an adaptive anti-tumoral immune response, characterised by an increase in naive B cells, follicular T helper cells, and activated NK cells; and another with a more undifferentiated response, with an increase in the activated-to-resting dendritic cells (DCs) ratio. Through a protein-protein interaction (PPI) network, we identified seven immunoregulatory hub genes: CD80, CD40, TNF, FCGR3A, CD163, FCGR3B, and CCR2. This study shows that, in Luminal A early-stage BC, SLNs gene expression studies enable the identification of distinct immune profiles that may influence prognosis stratification and highlight key genes that could serve as potential targets for immunotherapy.
Collapse
Affiliation(s)
- Joana Martins Ribeiro
- Laboratory of Sequencing and Functional Genomics of UCGenomics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - João Mendes
- Laboratory of Sequencing and Functional Genomics of UCGenomics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês Gante
- Gynecology Department, Coimbra Hospital and University Center, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal
- Gynecology University Clinic, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Margarida Figueiredo-Dias
- Gynecology Department, Coimbra Hospital and University Center, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal
- Gynecology University Clinic, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Vânia Almeida
- Department of Pathology, Coimbra Hospital and University Center, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Gomes
- Department of Pathology, Coimbra Hospital and University Center, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal
| | - Fernando Jesus Regateiro
- Laboratory of Sequencing and Functional Genomics of UCGenomics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Frederico Soares Regateiro
- Institute for Clinical and Biomedical Research (iCBR), Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Allergy and Clinical Immunology Unit, Coimbra Hospital and University Center, Unidade Local de Saúde de Coimbra, 3004-561 Coimbra, Portugal
- Institute of Immunology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Francisco Caramelo
- Institute for Clinical and Biomedical Research (iCBR), Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Laboratory of Biostatistics and Medical Informatics (LBIM), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
| | - Henriqueta Coimbra Silva
- Laboratory of Sequencing and Functional Genomics of UCGenomics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Institute for Clinical and Biomedical Research (iCBR), Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
| |
Collapse
|
7
|
Pazoki A, Dadfar S, Shadab A, Haghmorad D, Oksenych V. Soluble CD40 Ligand as a Promising Biomarker in Cancer Diagnosis. Cells 2024; 13:1267. [PMID: 39120299 PMCID: PMC11311304 DOI: 10.3390/cells13151267] [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: 06/27/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer remains a significant challenge in medicine due to its complexity and heterogeneity. Biomarkers have emerged as vital tools for cancer research and clinical practice, facilitating early detection, prognosis assessment, and treatment monitoring. Among these, CD40 ligand (CD40L) has gained attention for its role in immune response modulation. Soluble CD40 ligand (sCD40L) has shown promise as a potential biomarker in cancer diagnosis and progression, reflecting interactions between immune cells and the tumor microenvironment. This review explores the intricate relationship between sCD40L and cancer, highlighting its diagnostic and prognostic potential. It discusses biomarker discovery, emphasizing the need for reliable markers in oncology, and elucidates the roles of CD40L in inflammatory responses and interactions with tumor cells. Additionally, it examines sCD40L as a biomarker, detailing its significance across various cancer types and clinical applications. Moreover, the review focuses on therapeutic interventions targeting CD40L in malignancies, providing insights into cellular and gene therapy approaches and recombinant protein-based strategies. The clinical effectiveness of CD40L-targeted therapy is evaluated, underscoring the need for further research to unlock the full potential of this signaling pathway in cancer management.
Collapse
Affiliation(s)
- Alireza Pazoki
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Sepehr Dadfar
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Alireza Shadab
- Department of Health Science, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Dariush Haghmorad
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
- Cancer Research Center, Semnan University of Medical Sciences, Semnan 35147-99442, Iran
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
8
|
Zhang C, Singla RK, Tang M, Shen B. Natural products act as game-changer potentially in treatment and management of sepsis-mediated inflammation: A clinical perspective. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155710. [PMID: 38759311 DOI: 10.1016/j.phymed.2024.155710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 04/19/2024] [Accepted: 05/02/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Sepsis, a life-threatening condition resulting from uncontrolled host responses to infection, poses a global health challenge with limited therapeutic options. Due to high heterogeneity, sepsis lacks specific therapeutic drugs. Additionally, there remains a significant gap in the clinical management of sepsis regarding personalized and precise medicine. PURPOSE This review critically examines the scientific landscape surrounding natural products in sepsis and sepsis-mediated inflammation, highlighting their clinical potential. METHODS Following the PRISMA guidelines, we retrieved articles from PubMed to explore potential natural products with therapeutic effects in sepsis-mediated inflammation. RESULTS 434 relevant in vitro and in vivo studies were identified and screened. Ultimately, 55 studies were obtained as the supporting resources for the present review. We divided the 55 natural products into three categories: those influencing the synthesis of inflammatory factors, those affecting surface receptors and modulatory factors, and those influencing signaling pathways and the inflammatory cascade. CONCLUSION Natural products' potential as game-changers in sepsis-mediated inflammation management lies in their ability to modulate hallmarks in sepsis, including inflammation, immunity, and coagulopathy, which provides new therapeutic avenues that are readily accessible and capable of undergoing rapid clinical validation and deployment, offering a gift from nature to humanity. Innovative techniques like bioinformatics, metabolomics, and systems biology offer promising solutions to overcome these obstacles and facilitate the development of natural product-based therapeutics, holding promise for personalized and precise sepsis management and improving patient outcomes. However, standardization, bioavailability, and safety challenges arise during experimental validation and clinical trials of natural products.
Collapse
Affiliation(s)
- Chi Zhang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Min Tang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China; West China School of Nursing, Sichuan University, Chengdu, PR China
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610212, PR China.
| |
Collapse
|
9
|
Li MY, Ye W, Luo KW. Immunotherapies Targeting Tumor-Associated Macrophages (TAMs) in Cancer. Pharmaceutics 2024; 16:865. [PMID: 39065562 PMCID: PMC11280177 DOI: 10.3390/pharmaceutics16070865] [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: 05/21/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are one of the most plentiful immune compositions in the tumor microenvironment, which are further divided into anti-tumor M1 subtype and pro-tumor M2 subtype. Recent findings found that TAMs play a vital function in the regulation and progression of tumorigenesis. Moreover, TAMs promote tumor vascularization, and support the survival of tumor cells, causing an impact on tumor growth and patient prognosis. Numerous studies show that reducing the density of TAMs, or modulating the polarization of TAMs, can inhibit tumor growth, indicating that TAMs are a promising target for tumor immunotherapy. Recently, clinical trials have found that treatments targeting TAMs have achieved encouraging results, and the U.S. Food and Drug Administration has approved a number of drugs for use in cancer treatment. In this review, we summarize the origin, polarization, and function of TAMs, and emphasize the therapeutic strategies targeting TAMs in cancer treatment in clinical studies and scientific research, which demonstrate a broad prospect of TAMs-targeted therapies in tumor immunotherapy.
Collapse
Affiliation(s)
- Mei-Ye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
| | - Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
| | - Ke-Wang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; (M.-Y.L.); (W.Y.)
- People’s Hospital of Longhua, The affiliated hospital of Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
10
|
Abdulrehman T, Qadri S, Haik Y, Sultan A, Skariah S, Kumar S, Mendoza Z, Yadav KK, Titus A, Khader S. Advances in the targeted theragnostics of osteomyelitis caused by Staphylococcus aureus. Arch Microbiol 2024; 206:288. [PMID: 38834761 DOI: 10.1007/s00203-024-04015-2] [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: 03/31/2024] [Revised: 05/14/2024] [Accepted: 05/20/2024] [Indexed: 06/06/2024]
Abstract
Bone infections caused by Staphylococcus aureus may lead to an inflammatory condition called osteomyelitis, which results in progressive bone loss. Biofilm formation, intracellular survival, and the ability of S. aureus to evade the immune response result in recurrent and persistent infections that present significant challenges in treating osteomyelitis. Moreover, people with diabetes are prone to osteomyelitis due to their compromised immune system, and in life-threatening cases, this may lead to amputation of the affected limbs. In most cases, bone infections are localized; thus, early detection and targeted therapy may prove fruitful in treating S. aureus-related bone infections and preventing the spread of the infection. Specific S. aureus components or overexpressed tissue biomarkers in bone infections could be targeted to deliver active therapeutics, thereby reducing drug dosage and systemic toxicity. Compounds like peptides and antibodies can specifically bind to S. aureus or overexpressed disease markers and combining these with therapeutics or imaging agents can facilitate targeted delivery to the site of infection. The effectiveness of photodynamic therapy and hyperthermia therapy can be increased by the addition of targeting molecules to these therapies enabling site-specific therapy delivery. Strategies like host-directed therapy focus on modulating the host immune mechanisms or signaling pathways utilized by S. aureus for therapeutic efficacy. Targeted therapeutic strategies in conjunction with standard surgical care could be potential treatment strategies for S. aureus-associated osteomyelitis to overcome antibiotic resistance and disease recurrence. This review paper presents information about the targeting strategies and agents for the therapy and diagnostic imaging of S. aureus bone infections.
Collapse
Affiliation(s)
- Tahir Abdulrehman
- eHealth Program, DeGroote School of Business, McMaster University, Hamilton, ON, Canada
- Health Policy, Management and Informatics, Allied Health, Credit Valley Hospital, Mississauga, ON, Canada
| | - Shahnaz Qadri
- School of Pharmacy, Texas A&M University, Kingsville, USA.
| | - Yousef Haik
- Department of Mechanical & Nuclear Engineering, University of Sharjah, Sharjah, UAE.
| | - Ali Sultan
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Sini Skariah
- Department of Immunology & Microbiology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Shourya Kumar
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Zachary Mendoza
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Kamlesh K Yadav
- School of Engineering Medicine, Texas A&M University, Houston, TX, USA
| | - Anoop Titus
- Department of Preventive Cardiology, Houston Methodist, Houston, TX, USA
| | - Shameer Khader
- School of Public Health, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
11
|
Dakal TC, George N, Xu C, Suravajhala P, Kumar A. Predictive and Prognostic Relevance of Tumor-Infiltrating Immune Cells: Tailoring Personalized Treatments against Different Cancer Types. Cancers (Basel) 2024; 16:1626. [PMID: 38730579 PMCID: PMC11082991 DOI: 10.3390/cancers16091626] [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: 03/13/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
TIICs are critical components of the TME and are used to estimate prognostic and treatment responses in many malignancies. TIICs in the tumor microenvironment are assessed and quantified by categorizing immune cells into three subtypes: CD66b+ tumor-associated neutrophils (TANs), FoxP3+ regulatory T cells (Tregs), and CD163+ tumor-associated macrophages (TAMs). In addition, many cancers have tumor-infiltrating M1 and M2 macrophages, neutrophils (Neu), CD4+ T cells (T-helper), CD8+ T cells (T-cytotoxic), eosinophils, and mast cells. A variety of clinical treatments have linked tumor immune cell infiltration (ICI) to immunotherapy receptivity and prognosis. To improve the therapeutic effectiveness of immune-modulating drugs in a wider cancer patient population, immune cells and their interactions in the TME must be better understood. This study examines the clinicopathological effects of TIICs in overcoming tumor-mediated immunosuppression to boost antitumor immune responses and improve cancer prognosis. We successfully analyzed the predictive and prognostic usefulness of TIICs alongside TMB and ICI scores to identify cancer's varied immune landscapes. Traditionally, immune cell infiltration was quantified using flow cytometry, immunohistochemistry, gene set enrichment analysis (GSEA), CIBERSORT, ESTIMATE, and other platforms that use integrated immune gene sets from previously published studies. We have also thoroughly examined traditional limitations and newly created unsupervised clustering and deconvolution techniques (SpatialVizScore and ProTICS). These methods predict patient outcomes and treatment responses better. These models may also identify individuals who may benefit more from adjuvant or neoadjuvant treatment. Overall, we think that the significant contribution of TIICs in cancer will greatly benefit postoperative follow-up, therapy, interventions, and informed choices on customized cancer medicines.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur 313001, Rajasthan, India
| | - Nancy George
- Department of Biotechnology, Chandigarh University, Mohali 140413, Punjab, India;
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of the City of Hope, Monrovia, CA 91010, USA;
| | - Prashanth Suravajhala
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Clappana P.O. 690525, Kerala, India;
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, Karnataka, India
| |
Collapse
|
12
|
Zhou Y, Richmond A, Yan C. Harnessing the potential of CD40 agonism in cancer therapy. Cytokine Growth Factor Rev 2024; 75:40-56. [PMID: 38102001 PMCID: PMC10922420 DOI: 10.1016/j.cytogfr.2023.11.002] [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: 11/02/2023] [Accepted: 11/22/2023] [Indexed: 12/17/2023]
Abstract
CD40 is a member of the tumor necrosis factor (TNF) receptor superfamily of receptors expressed on a variety of cell types. The CD40-CD40L interaction gives rise to many immune events, including the licensing of dendritic cells to activate CD8+ effector T cells, as well as the facilitation of B cell activation, proliferation, and differentiation. In malignant cells, the expression of CD40 varies among cancer types, mediating cellular proliferation, apoptosis, survival and the secretion of cytokines and chemokines. Agonistic human anti-CD40 antibodies are emerging as an option for cancer treatment, and early-phase clinical trials explored its monotherapy or combination with radiotherapy, chemotherapy, immune checkpoint blockade, and other immunomodulatory approaches. In this review, we present the current understanding of the mechanism of action for CD40, along with results from the clinical development of agonistic human CD40 antibodies in cancer treatment (selicrelumab, CDX-1140, APX005M, mitazalimab, 2141-V11, SEA-CD40, LVGN7409, and bispecific antibodies). This review also examines the safety profile of CD40 agonists in both preclinical and clinical settings, highlighting optimized dosage levels, potential adverse effects, and strategies to mitigate them.
Collapse
Affiliation(s)
- Yang Zhou
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA
| | - Chi Yan
- Tennessee Valley Healthcare System, Department of Veteran Affairs, Nashville, TN, USA; Vanderbilt University School of Medicine, Department of Pharmacology, Nashville, TN, USA.
| |
Collapse
|
13
|
Ding H, An G. Role of the CD40-CD40L expression level pathway in the diagnosis of unexplained recurrent pregnancy loss. J OBSTET GYNAECOL 2023; 43:2280840. [PMID: 38035611 DOI: 10.1080/01443615.2023.2280840] [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: 01/09/2023] [Accepted: 11/03/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Unexplained recurrent spontaneous pregnancy loss (URPL) lacks effective treatment and reliable early diagnosis and prediction. Immunologic dysfunction can be an underlying cause of recurrent pregnancy loss (RPL). Considering the regulatory role of CD40-CD40L in immune responses, we explored its clinical significance in URPL. METHODS The 108 women with URPL who were treated in Hebei Yanda Hospital from January 2020 to December 2022 were selected as study subjects, and another 108 healthy women who were not pregnant and matched with the age and body mass index of the study group were selected as the control group. CD40 and CD4 + CD25 + Treg cells in peripheral blood mononuclear cells (PBMCs) and CD40L in peripheral blood platelets were measured by flow cytometry. The predictive value of CD40-CD40L in URPL for the risk of RPL was determined by receiver operating characteristic (ROC) curves. The correlations of CD40-CD40L with CD4 + CD25 + Treg cells and serum pro-inflammatory factors were assessed by Pearson's analysis. RESULTS CD40 on the surface of PBMCs and CD40L on the surface of platelets were up-regulated in URPL patients. CD40 in combination with CD40L had high predictive value for the risk of RPL in URPL patients. Peripheral blood CD40-CD40L was positively linked to IL-17 and IL-23, and negatively to CD4 + CD25 + Treg cells and IL-10 in URPL patients. CONCLUSIONS The CD40-CD40L pathway expression in peripheral blood can help predict the risk of RPL in URPL patients.
Collapse
Affiliation(s)
- Hui Ding
- Department of Obstetrics and Gynecology, Hebei Yanda Hospital, Langfang City, China
| | - Guoqian An
- Department of Obstetrics and Gynecology, Hebei Yanda Hospital, Langfang City, China
| |
Collapse
|
14
|
Thakur M, Singh M, Kumar S, Dwivedi VP, Dakal TC, Yadav V. A Reappraisal of the Antiviral Properties of and Immune Regulation through Dietary Phytochemicals. ACS Pharmacol Transl Sci 2023; 6:1600-1615. [PMID: 37974620 PMCID: PMC10644413 DOI: 10.1021/acsptsci.3c00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Indexed: 11/19/2023]
Abstract
In the present era of the COVID-19 pandemic, viral infections remain a major cause of morbidity and mortality worldwide. In this day and age, viral infections are rampant and spreading rapidly. Among the most aggressive viral infections are ebola, AIDS (acquired immunodeficiency syndrome), influenza, and SARS (severe acute respiratory syndrome). Even though there are few treatment options for viral diseases, most of the antiviral therapies are ineffective owing to frequent mutations, the development of more aggressive strains, drug resistance, and possible side effects. Traditionally, herbal remedies have been used by healers, including for dietary and medicinal purposes. Many clinical and scientific studies have demonstrated the therapeutic potential of plant-derived natural compounds. Because of unsafe practices like blood transfusions and organ transplants from infected patients, medical supply contamination. Our antiviral therapies cannot achieve sterile immunity, and we have yet to find a cure for these pernicious infections. Herbs have been shown to improve therapeutic efficacy against a wide variety of viral diseases because of their high concentration of immunomodulatory phytochemicals (both immunoinhibitory and anti-inflammatory). Combined with biotechnology, this folk medicine system can lead to the development of novel antiviral drugs and therapies. In this Review, we will summarize some selected bioactive compounds with probable mechanisms of their antiviral actions, focusing on the immunological axis of these compounds.
Collapse
Affiliation(s)
- Mony Thakur
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Mona Singh
- Department
of Obstetrics and Gynaecology, Medical College
of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Sandeep Kumar
- Division
of Cell Biology and Immunology, Council
of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh 160036, India
| | - Ved Prakash Dwivedi
- International
Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Tikam Chand Dakal
- Genome
and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India
| | - Vinod Yadav
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| |
Collapse
|
15
|
Krykbaeva I, Bridges K, Damsky W, Pizzurro GA, Alexander AF, McGeary MK, Park K, Muthusamy V, Eyles J, Luheshi N, Turner N, Weiss SA, Olino K, Kaech SM, Kluger HM, Miller-Jensen K, Bosenberg M. Combinatorial Immunotherapy with Agonistic CD40 Activates Dendritic Cells to Express IL12 and Overcomes PD-1 Resistance. Cancer Immunol Res 2023; 11:1332-1350. [PMID: 37478171 DOI: 10.1158/2326-6066.cir-22-0699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/17/2023] [Accepted: 07/20/2023] [Indexed: 07/23/2023]
Abstract
Checkpoint inhibitors have revolutionized cancer treatment, but resistance remains a significant clinical challenge. Myeloid cells within the tumor microenvironment can modulate checkpoint resistance by either supporting or suppressing adaptive immune responses. Using an anti-PD-1-resistant mouse melanoma model, we show that targeting the myeloid compartment via CD40 activation and CSF1R blockade in combination with anti-PD-1 results in complete tumor regression in a majority of mice. This triple therapy combination was primarily CD40 agonist-driven in the first 24 hours after therapy and showed a similar systemic cytokine profile in human patients as was seen in mice. Functional single-cell cytokine secretion profiling of dendritic cells (DC) using a novel microwell assay identified a CCL22+CCL5+ IL12-secreting DC subset as important early-stage effectors of triple therapy. CD4+ and CD8+ T cells are both critical effectors of treatment, and systems analysis of single-cell RNA sequencing data supported a role for DC-secreted IL12 in priming T-cell activation and recruitment. Finally, we showed that treatment with a novel IL12 mRNA therapeutic alone was sufficient to overcome PD-1 resistance and cause tumor regression. Overall, we conclude that combining myeloid-based innate immune activation and enhancement of adaptive immunity is a viable strategy to overcome anti-PD-1 resistance.
Collapse
Affiliation(s)
- Irina Krykbaeva
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Kate Bridges
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - William Damsky
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Gabriela A Pizzurro
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Amanda F Alexander
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Meaghan K McGeary
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Koonam Park
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Viswanathan Muthusamy
- Yale Center for Precision Cancer Modeling, Yale School of Medicine, New Haven, Connecticut
| | - James Eyles
- Oncology Research and Early Development, AstraZeneca, Cambridge, United Kingdom
| | - Nadia Luheshi
- Oncology Research and Early Development, AstraZeneca, Cambridge, United Kingdom
| | - Noel Turner
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Sarah A Weiss
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kelly Olino
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute of Biological Sciences, La Jolla, California
| | - Harriet M Kluger
- Department of Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Kathryn Miller-Jensen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut
| | - Marcus Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
- Yale Center for Precision Cancer Modeling, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
16
|
Yu D, Yang P, Lu X, Huang S, Liu L, Fan X. Single-cell RNA sequencing reveals enhanced antitumor immunity after combined application of PD-1 inhibitor and Shenmai injection in non-small cell lung cancer. Cell Commun Signal 2023; 21:169. [PMID: 37430270 DOI: 10.1186/s12964-023-01184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 06/04/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have altered the clinical management of non-small cell lung cancer (NSCLC). However, the low response rate, severe immune-related adverse events (irAEs), and hyperprogressive disease following ICIs monotherapy require attention. Combination therapy may overcome these limitations and traditional Chinese medicine with immunomodulatory effects provides a promising approach. Shenmai injection (SMI) is a clinically effective adjuvant treatment for cancer with chemotherapy and radiotherapy. Therefore, the combined effects and mechanisms of SMI and programmed death-1 (PD-1) inhibitor against NSCLC was focused on this study. METHODS A Lewis lung carcinoma mouse model and a lung squamous cell carcinoma humanized mouse model were used to investigate the combined efficacy and safety of SMI and PD-1 inhibitor. The synergistic mechanisms of the combination therapy against NSCLC were explored using single-cell RNA sequencing. Validation experiments were performed using immunofluorescence analysis, in vitro experiment, and bulk transcriptomic datasets. RESULTS In both models, combination therapy alleviated tumor growth and prolonged survival without increasing irAEs. The GZMAhigh and XCL1high natural killer (NK) cell subclusters with cytotoxic and chemokine signatures increased in the combination therapy, while malignant cells from combination therapy were mainly in the apoptotic state, suggesting that mediating tumor cell apoptosis through NK cells is the main synergistic mechanisms of combination therapy. In vitro experiment confirmed that combination therapy increased secretion of Granzyme A by NK cells. Moreover, we discovered that PD-1 inhibitor and SMI combination blocked inhibitory receptors on NK and T cells and restores their antitumoral activity in NSCLC better than PD-1 inhibitor monotherapy, and immune and stromal cells exhibited a decrease of angiogenic features and attenuated cancer metabolism reprogramming in microenvironment of combination therapy. CONCLUSIONS This study demonstrated that SMI reprograms tumor immune microenvironment mainly by inducing NK cells infiltration and synergizes with PD-1 inhibitor against NSCLC, suggested that targeting NK cells may be an important strategy for combining with ICIs. Video Abstract.
Collapse
Affiliation(s)
- Dingyi Yu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Penghui Yang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Shaoze Huang
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China
| | - Li Liu
- Zhejiang Engineering Research Center for Advanced Manufacturing of Traditional Chinese Medicine, Huzhou, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
- Innovation Center in Zhejiang University, State Key Laboratory of Component-Based Chinese Medicine, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
17
|
Zhang Q, Liu X, Pang X, Wang H, Du J, Ren H, Hu P. Recovery of host adaptive immune function promoted the reduction of hepatitis B surface antigen in nucleoside analog-experienced chronic hepatitis B patients with low hepatitis B surface antigen levels. Cytokine 2023; 164:156140. [PMID: 36738524 DOI: 10.1016/j.cyto.2023.156140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/21/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023]
Abstract
Hepatitis B surface antigen (HBsAg) seroclearance is an optimal therapeutic endpoint, as it reflects the amount of covalently closed circular DNA. The exact mechanisms that contribute to HBsAg reduction are not completely understood. We evaluated adaptive immunity in nucleoside analog-experienced chronic hepatitis B (CHB) patients with low HBsAg levels who received oral antiviral therapy. One hundred and ninety-five CHB patients had hepatitis B virus (HBV) DNA ≤ 1000 IU/ml and HBsAg < 3000 IU/ml for longer than one year of antiviral therapy. According to HBsAg levels, they were divided into Group 1 (HBsAg reduction ≥ 0.5 log10) and Group 2 (HBsAg reduction < 0.5 log10). Cytokines, adaptive immune cells, and molecular markers in peripheral blood were detected at follow-up times. In total, 38 (19.5%) of the 195 patients achieved HBsAg reduction ≥ 0.5 log10. IL4, IL5, IL10, TGF β, IL17, and PD-1 decreased gradually in these patients. HBsAg reduction had a link to the change in ICOSL+CD19+ B cells and CD40L+CXCR5+CD4+ Tfh cells. More CD8+ naive T lymphocytes differentiated into CD4+ TCMs, CD8+ TCMs and CD8+ TEMs in Group 1. Meanwhile, Group 1 exhibited elevated Th1 and Th1/Th2 levels and reduced levels of Treg versus those in Group 2. With the reduction in HBsAg, the imbalance of T-cell subsets was partially corrected; the immune activity of T cells was enhanced, and the state of immune exhaustion was alleviated to a certain extent.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, China; Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqing Liu
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xicheng Pang
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huimin Wang
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinjie Du
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Ren
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Hu
- Department of Infectious Diseases, Institute for Viral Hepatitis, The Key Laboratory of Molecular Biology for Infectious Diseases, Chinese Ministry of Education, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
18
|
Vargas-Uricoechea H. Molecular Mechanisms in Autoimmune Thyroid Disease. Cells 2023; 12:918. [PMID: 36980259 PMCID: PMC10047067 DOI: 10.3390/cells12060918] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
The most common cause of acquired thyroid dysfunction is autoimmune thyroid disease, which is an organ-specific autoimmune disease with two presentation phenotypes: hyperthyroidism (Graves-Basedow disease) and hypothyroidism (Hashimoto's thyroiditis). Hashimoto's thyroiditis is distinguished by the presence of autoantibodies against thyroid peroxidase and thyroglobulin. Meanwhile, autoantibodies against the TSH receptor have been found in Graves-Basedow disease. Numerous susceptibility genes, as well as epigenetic and environmental factors, contribute to the pathogenesis of both diseases. This review summarizes the most common genetic, epigenetic, and environmental mechanisms involved in autoimmune thyroid disease.
Collapse
Affiliation(s)
- Hernando Vargas-Uricoechea
- Metabolic Diseases Study Group, Department of Internal Medicine, Universidad del Cauca, Carrera 6 Nº 13N-50, Popayán 190001, Colombia
| |
Collapse
|
19
|
Zheng C, Shi Y, Zou Y. T cell co-stimulatory and co-inhibitory pathways in atopic dermatitis. Front Immunol 2023; 14:1081999. [PMID: 36993982 PMCID: PMC10040887 DOI: 10.3389/fimmu.2023.1081999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/28/2023] [Indexed: 03/14/2023] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) targeting the T cell inhibitory pathways has revolutionized cancer treatment. However, ICIs might induce progressive atopic dermatitis (AD) by affecting T cell reactivation. The critical role of T cells in AD pathogenesis is widely known. T cell co-signaling pathways regulate T cell activation, where co-signaling molecules are essential for determining the magnitude of the T cell response to antigens. Given the increasing use of ICIs in cancer treatment, a timely overview of the role of T cell co-signaling molecules in AD is required. In this review, we emphasize the importance of these molecules involved in AD pathogenesis. We also discuss the potential of targeting T cell co-signaling pathways to treat AD and present the unresolved issues and existing limitations. A better understanding of the T cell co-signaling pathways would aid investigation of the mechanism, prognosis evaluation, and treatment of AD.
Collapse
Affiliation(s)
- Chunjiao Zheng
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Institute of Psoriasis, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| | - Ying Zou
- Skin and Cosmetic Research Department, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Yuling Shi, ; Ying Zou,
| |
Collapse
|
20
|
Chattopadhyay P, Khare K, Kumar M, Mishra P, Anand A, Maurya R, Gupta R, Sahni S, Gupta A, Wadhwa S, Yadav A, Devi P, Tardalkar K, Joshi M, Sethi T, Pandey R. Single-cell multiomics revealed the dynamics of antigen presentation, immune response and T cell activation in the COVID-19 positive and recovered individuals. Front Immunol 2022; 13:1034159. [PMID: 36532041 PMCID: PMC9755500 DOI: 10.3389/fimmu.2022.1034159] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction Despite numerous efforts to describe COVID-19's immunological landscape, there is still a gap in our understanding of the virus's infections after-effects, especially in the recovered patients. This would be important to understand as we now have huge number of global populations infected by the SARS-CoV-2 as well as variables inclusive of VOCs, reinfections, and vaccination breakthroughs. Furthermore, single-cell transcriptome alone is often insufficient to understand the complex human host immune landscape underlying differential disease severity and clinical outcome. Methods By combining single-cell multi-omics (Whole Transcriptome Analysis plus Antibody-seq) and machine learning-based analysis, we aim to better understand the functional aspects of cellular and immunological heterogeneity in the COVID-19 positive, recovered and the healthy individuals. Results Based on single-cell transcriptome and surface marker study of 163,197 cells (124,726 cells after data QC) from the 33 individuals (healthy=4, COVID-19 positive=16, and COVID-19 recovered=13), we observed a reduced MHC Class-I-mediated antigen presentation and dysregulated MHC Class-II-mediated antigen presentation in the COVID-19 patients, with restoration of the process in the recovered individuals. B-cell maturation process was also impaired in the positive and the recovered individuals. Importantly, we discovered that a subset of the naive T-cells from the healthy individuals were absent from the recovered individuals, suggesting a post-infection inflammatory stage. Both COVID-19 positive patients and the recovered individuals exhibited a CD40-CD40LG-mediated inflammatory response in the monocytes and T-cell subsets. T-cells, NK-cells, and monocyte-mediated elevation of immunological, stress and antiviral responses were also seen in the COVID-19 positive and the recovered individuals, along with an abnormal T-cell activation, inflammatory response, and faster cellular transition of T cell subtypes in the COVID-19 patients. Importantly, above immune findings were used for a Bayesian network model, which significantly revealed FOS, CXCL8, IL1β, CST3, PSAP, CD45 and CD74 as COVID-19 severity predictors. Discussion In conclusion, COVID-19 recovered individuals exhibited a hyper-activated inflammatory response with the loss of B cell maturation, suggesting an impeded post-infection stage, necessitating further research to delineate the dynamic immune response associated with the COVID-19. To our knowledge this is first multi-omic study trying to understand the differential and dynamic immune response underlying the sample subtypes.
Collapse
Affiliation(s)
- Partha Chattopadhyay
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kriti Khare
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manish Kumar
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Pallavi Mishra
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Alok Anand
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Ranjeet Maurya
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rohit Gupta
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Shweta Sahni
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Ayushi Gupta
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Saruchi Wadhwa
- CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
| | - Aanchal Yadav
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Priti Devi
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kishore Tardalkar
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Meghnad Joshi
- Department of Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Medical College, Hospital and Research Institute, Kolhapur, Maharashtra, India
| | - Tavpritesh Sethi
- Indraprastha Institute of Information Technology Delhi, New Delhi, India
| | - Rajesh Pandey
- Division of Immunology and Infectious Disease Biology, INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
21
|
Wu J, Wang Y. Role of TNFSF9 bidirectional signal transduction in antitumor immunotherapy. Eur J Pharmacol 2022; 928:175097. [PMID: 35714694 DOI: 10.1016/j.ejphar.2022.175097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022]
Abstract
The complex structure of the tumor microenvironment leads to the poor efficacy of tumor immunotherapy. The therapeutic adjuvant designed to enhance the effect of T cells by acting on the costimulatory molecule tumor necrosis factor superfamily member 9 (TNFSF9) has achieved good results. However, because some tumors are characterized by reduced T-cell infiltration, adjuvants acting on T cells alone may have limitations. On the other hand, the blockade of TNFSF9 reverse signalling can have an antitumor effect by reshaping the tumor microenvironment. Therefore, this paper mainly discusses the current status and potential of TNFSF9 bidirectional signalling in antitumor immunotherapy to provide new ideas for tumor immunotherapy.
Collapse
Affiliation(s)
- Jiao Wu
- Departments of Gastroenterology, Mianyang Central Hospital, Sichuan, 621000, China
| | - Yunpeng Wang
- Departments of Cardiology, Mianyang Central Hospital, Sichuan, 621000, China.
| |
Collapse
|
22
|
Hu XX, Zhang AJ, Pan WW, Xin QL, Chen JY, Zhang LL, Chang Y, Wu YJ, Wei W. An IgD-Fc-Ig fusion protein restrains the activation of T and B cells by inhibiting IgD-IgDR-Lck signaling in rheumatoid arthritis. Acta Pharmacol Sin 2022; 43:387-400. [PMID: 33864023 PMCID: PMC8791948 DOI: 10.1038/s41401-021-00665-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/19/2021] [Indexed: 02/08/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by synovitis and the destruction of small joints. Emerging evidence shows that immunoglobulin D (IgD) stimulation induces T-cell activation, which may contribute to diseases pathogenesis in RA. In this study, we investigated the downstream signaling pathways by which IgD activated T cells as well as the possible role of IgD in the T-B interaction. Peripheral blood mononuclear cells were isolated from peripheral blood of healthy controls and RA patients. We demonstrated that IgD activated T cells through IgD receptor (IgDR)-lymphocyte-specific protein tyrosine kinase (Lck)-zeta-associated protein 70 (ZAP70)/phosphatidylinositol 3-kinase (PI3K)/nuclear factor kappa-B (NF-κB) signaling pathways; IgD-induced CD4+ T cells promoted the proliferation of CD19+ B cells in RA patients. A novel fusion protein IgD-Fc-Ig (composed of human IgD-Fc domain and IgG1 Fc domain, which specifically blocked the IgD-IgDR binding) inhibited the coexpression of IgDR and phosphorylated Lck (p-Lck) and the expression levels of p-Lck, p-ZAP70, p-PI3K on CD4+ T cells, and decreased NF-κB nuclear translocation in Jurkat cells. Meanwhile, IgD-Fc-Ig downregulated the expression levels of CD40L on CD4+ T cells as well as CD40, CD86 on CD19+ B cells in RA patients and healthy controls. It also decreased the expression levels of CD40L on CD4+ T cells and CD40 on CD19+ B cells from spleens of collagen-induced arthritis (CIA) mice and reduced IL-17A level in mouse serum. Moreover, administration of IgD-Fc-Ig (1.625-13 mg/kg, iv, twice a week for 4 weeks) in CIA mice dose-dependently decreased the protein expression levels of CD40, CD40L, and IgD in spleens. IgD-Fc-Ig restrains T-cell activation through inhibiting IgD-IgDR-Lck-ZAP70-PI3K-NF-κB signaling, thus inhibiting B-cell activation. Our data provide experimental evidences for application of IgD-Fc-Ig as a highly selective T cell-targeting treatment for RA.
Collapse
Affiliation(s)
- Xiao-xi Hu
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Ai-jun Zhang
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Wen-wen Pan
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Qian-ling Xin
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Jing-yu Chen
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Ling-ling Zhang
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Yan Chang
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Yu-jing Wu
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| | - Wei Wei
- grid.186775.a0000 0000 9490 772XInstitute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, 230032 China
| |
Collapse
|
23
|
Lin Y, Liu Q, Chen Z, Zheng F, Huang H, Yu C, Yang J. The immunomodulatory effect of koumine on B cells under dependent and independent responses by T cells. Eur J Pharmacol 2022; 914:174690. [PMID: 34890543 DOI: 10.1016/j.ejphar.2021.174690] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 11/03/2022]
Abstract
Dysregulated activation of polyclonal B cells and production of pathogenic antibodies are involved in the development of rheumatoid arthritis (RA). Therefore, targeted B cell therapy is effective against RA. Gelsemium elegans (Gardn. & Champ.) Benth., a toxic plant widely distributed in Southeast Asia, has been used for treating rheumatoid pain, neuropathic pain, spasticity, skin ulcers, and cancers for many years in traditional Chinese medicine. Koumine, an alkaloid monomer from Gelsemium elegans Benth., exerts therapeutic effects against RA. However, whether koumine affects B cells remains unknown. In this study, the effect of koumine on B cells under T cell-independent (TI) and T cell-dependent (TD) immune responses is investigated in vitro and in vivo. Mouse primary B cells were obtained by immunomagnetic bead sorting, and immunomodulatory effects of koumine on the activation, proliferation, and differentiation of B cells were determined in TI and TD models induced by lipopolysaccharide (LPS) and anti-CD40 antibodies in vitro, respectively. The humoral immune responses of TI and TD were established using NP-AECM-FICOLL and NP-CGG in C57BL/6J mice, respectively. We found that koumine inhibited B cell differentiation in the TI model and inhibited B cell activation and proliferation in the TD model in vitro. Koumine also inhibited antibody secretion in TI immune response, TD initial immune response, and in TD secondary immune response. Our results reveal that koumine has a direct and indirect immune regulatory effect on B cells, showing that it can directly inhibit the differentiation and secretion of autoantibodies after abnormal activation of B cells, and indirectly inhibit the activation and proliferation of TD B cells to reduce the secretion of antibodies. It may be an important mechanism for its anti-RA effect in mice, providing a rationale and laboratory data support for the application of koumine in anti-human RA therapy.
Collapse
Affiliation(s)
- Yarong Lin
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Qian Liu
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Zehong Chen
- Laboratory of Medical Function, Basic Medical Experimental Teaching Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Fengting Zheng
- Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Huihui Huang
- Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Changxi Yu
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Drug Target Discovery and Structural and Functional Research, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Jian Yang
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; Fujian Key Laboratory of Natural Medicine Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| |
Collapse
|
24
|
Yan C, Richmond A. Hiding in the dark: pan-cancer characterization of expression and clinical relevance of CD40 to immune checkpoint blockade therapy. Mol Cancer 2021; 20:146. [PMID: 34758832 PMCID: PMC8582157 DOI: 10.1186/s12943-021-01442-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/04/2021] [Indexed: 11/10/2022] Open
Abstract
HIGHLIGHTS CD40 expression correlates with the type I anti-tumor response and better survival. Pan-cancer bioinformatics characterization reveals reduced CD40 expression in 11 cancer types, including RASmut melanoma compared to nevi. RAS mutation correlates with reduced CD40 expression in malignant melanoma. CD40 expression is associated with better response to immune checkpoint blockade therapy in melanoma.
Collapse
Affiliation(s)
- Chi Yan
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA.,Department of Pharmacology, Vanderbilt University School of Medicine, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, TN, USA. .,Department of Pharmacology, Vanderbilt University School of Medicine, 432 PRB, 2220 Pierce Ave, Nashville, TN, 37232, USA.
| |
Collapse
|
25
|
Simultaneous Inhibition of PD-1 and Stimulation of CD40 Signaling Pathways by Anti-PD-L1/CD40L Bispecific Fusion Protein Synergistically Activate Target and Effector Cells. Int J Mol Sci 2021; 22:ijms222111302. [PMID: 34768776 PMCID: PMC8583728 DOI: 10.3390/ijms222111302] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/22/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Bispecific antibodies (BsAbs) or fusion proteins (BsAbFPs) present a promising strategy for cancer immunotherapy. Numerous BsAbs targeting coinhibitory and costimulatory pathways have been developed for retargeting T cells and antigen presenting cells (APCs). It is challenging to assess the potency of BsAb that engages two different signaling pathways simultaneously in a single assay format, especially when the two antigen targets are expressed on different cells. To explore the potency of anti-PD-L1/CD40L BsAbFP, a fusion protein that binds to human CD40 and PD-L1, we engineered CHO cells as surrogate APCs that express T cell receptor activator and PD-L1, Jurkat cells with PD-1 and NFAT-luciferase reporter as effector T cells, and Raji cell with NFkB-luciferase that endogenously expresses CD40 as accessory B cells. A novel reporter gene bioassay was developed using these cell lines that allows anti-PD-L1/CD40L BsAbFP to engages both PD-1/PD-L1 and CD40/CD40L signaling pathways in one assay. As both reporters use firefly luciferase, the effects of activating both signaling pathways is observed as an increase in luminescence, either as a higher upper asymptote, a lower EC50, or both. This dual target reporter gene bioassay system reflects potential mechanism of action and demonstrated the ability of anti-PD-L1/CD40L BsAbFP to synergistically induce biological response compared to the combination of anti-PD-L1 monovalent monoclonal antibody and agonist CD40L fusion protein, or either treatment alone. The results also showed a strong correlation between the drug dose and biological response within the tested potency range with good linearity, accuracy, precision, specificity and stability indicating properties, suggesting that this “three-cell-in-one” dual target reporter gene bioassay is suitable for assessing potency, structure-function and critical quality attributes of anti-PD-L1/CD40L BsAbFP. This approach could be used for developing dual target bioassays for other BsAbs and antibodies used for combination therapy.
Collapse
|
26
|
Clinical Implementation of Biologics and Small Molecules in the Treatment of Hidradenitis Suppurativa. Drugs 2021; 81:1397-1410. [PMID: 34283386 PMCID: PMC8352818 DOI: 10.1007/s40265-021-01566-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2021] [Indexed: 01/03/2023]
Abstract
Hidradenitis suppurativa (HS) is a chronic, recurrent, auto-inflammatory skin disease originating from the hair follicles. The typical inflammatory nodules, abscesses, and draining sinus tracts (tunnels) are characterized by a massive influx of neutrophils, macrophages, B-cells, plasma cells, T helper (Th)1, Th17 cells and upregulation of pro-inflammatory cytokines such as IL-1, IL-17, IL-12/23, and TNF-α. Over the last decades, several clinical trials evaluated the clinical efficacy of different biologics targeting these pro-inflammatory cytokines, in particular TNF-α and IL-1. However, adalimumab is still the only registered drug for HS. This review discusses biologics and small molecules with high level of evidence for their clinical application, provides guidance on when and how to use these biologics and small molecules in clinical practice, and elaborates on the combination with medical and surgical treatment options beyond the current guidelines. Furthermore this review provides an overview of potential biologics and small molecules currently under investigation for novel targets in HS such as IL-36, C5a, Janus kinase family members, CD-40, LTA4 and CXCR1/2.
Collapse
|
27
|
Liu S, Xu J, Wu J. The Role of Co-Signaling Molecules in Psoriasis and Their Implications for Targeted Treatment. Front Pharmacol 2021; 12:717042. [PMID: 34354596 PMCID: PMC8329336 DOI: 10.3389/fphar.2021.717042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Psoriasis is a chronic, systemic immune-mediated inflammatory disease manifesting in the skin, joint or both. Co-signaling molecules are essential for determining the magnitude of the T cell response to the antigen. According to the function of co-signaling molecules, they can be divided into co-stimulatory molecules and co-inhibitory molecules. The role of co-signaling molecules in psoriasis is recognized, mainly including the co-stimulatory molecules CD28, CD40, OX40, CD27, DR3, LFA-1, and LFA-3 and the co-inhibitory molecules CTLA-4, PD-1, and TIM-3. They impact the pathological process of psoriasis by modulating the immune strength of T cells, regulating the production of cytokines or the differentiation of Tregs. In recent years, immunotherapies targeting co-signaling molecules have made significant progress and shown broad application prospects in psoriasis. This review aims to outline the possible role of co-signaling molecules in the pathogenesis of psoriasis and their potential application for the treatment of psoriasis.
Collapse
Affiliation(s)
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Agonistic CD40 Antibodies in Cancer Treatment. Cancers (Basel) 2021; 13:cancers13061302. [PMID: 33804039 PMCID: PMC8000216 DOI: 10.3390/cancers13061302] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/16/2021] [Accepted: 03/12/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary CD40 is a costimulatory molecule that is key for the activation of antigen-presenting cells and other innate immune cells. It plays an important role in anti-tumor immunity, and agonists of CD40 have been shown to eliminate tumors in both pre-clinical and clinical settings, alone and in combination with other treatment modalities. Here we assess the expression of CD40 and associations with other mediators of immunity in a variety of tumor types and review the potential of CD40 agonists for cancer treatment, given the promise of enhancing the interplay between innate and adaptive immunity. Abstract CD40 is expressed on a variety of antigen-presenting cells. Stimulation of CD40 results in inflammation by upregulation of other costimulatory molecules, increased antigen presentation, maturation (licensing) of dendritic cells, and activation of CD8+ T cells. Here we analyzed gene expression data from The Cancer Genome Atlas in melanoma, renal cell carcinoma, and pancreatic adenocarcinoma and found correlations between CD40 and several genes involved in antigen presentation and T cell function, supporting further exploration of CD40 agonists to treat cancer. Agonist CD40 antibodies have induced anti-tumor effects in several tumor models and the effect has been more pronounced when used in combination with other treatments (immune checkpoint inhibition, chemotherapy, and colony-stimulating factor 1 receptor inhibition). The reduction in tumor growth and ability to reprogram the tumor microenvironment in preclinical models lays the foundation for clinical development of agonistic CD40 antibodies (APX005M, ChiLob7/4, ADC-1013, SEA-CD40, selicrelumab, and CDX-1140) that are currently being evaluated in early phase clinical trials. In this article, we focus on CD40 expression and immunity in cancer, agonistic human CD40 antibodies, and their pre-clinical and clinical development. With the broad pro-inflammatory effects of CD40 and its ligand on dendritic cells and macrophages, and downstream B and T cell activation, agonists of this pathway may enhance the anti-tumor activity of other systemic therapies.
Collapse
|
29
|
Sharma A, Liu H, Herwig-Carl MC, Chand Dakal T, Schmidt-Wolf IGH. Epigenetic Regulatory Enzymes: mutation Prevalence and Coexistence in Cancers. Cancer Invest 2021; 39:257-273. [PMID: 33411587 DOI: 10.1080/07357907.2021.1872593] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epigenetic regulation is an important layer of transcriptional control with the particularity to affect the broad spectrum of genome. Over the years, largely due to the substantial number of recurrent mutations, there have been hundreds of novel driver genes characterized in various cancers. Additionally, the relative contribution of two dysregulated epigenomic entities (DNA methylation and histone modifications) that gradually drive the cancer phenotype remains in the research focus. However, a complex scenario arises when the disease phenotype does not harbor any relevant mutation or an abnormal transcription level. Although the cancer landscape involves the contribution of multiple genetic and non-genetic factors, herein, we discuss specifically the mutation spectrum of epigenetically-related enzymes in cancer. In addition, we address the coexistence of these two epigenetic entities in malignant human diseases, especially cancer. We suggest that the study of epigenetically-related somatic mutations in the early cellular differentiation stage of embryonic development might help to understand their later-staged footprints in the cancer genome. Furthermore, understanding the co-occurrence and/or inverse association of different disease types and redefining the general definition of "healthy" controls could provide insights into the genome reorganization.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Bonn, Germany.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | | | - Tikam Chand Dakal
- Department of Biotechnology, Mohanlal Sukhadia University, Rajasthan, India
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, CIO Bonn, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
30
|
Monteiro de Oliveira Novaes JA, Hirz T, Guijarro I, Nilsson M, Pisegna MA, Poteete A, Barsoumian HB, Fradette JJ, Chen LN, Gibbons DL, Tian X, Wang J, Myers JN, McArthur MJ, Bell D, William WN, Heymach JV. Targeting of CD40 and PD-L1 Pathways Inhibits Progression of Oral Premalignant Lesions in a Carcinogen-induced Model of Oral Squamous Cell Carcinoma. Cancer Prev Res (Phila) 2020; 14:313-324. [PMID: 33277316 DOI: 10.1158/1940-6207.capr-20-0418] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/30/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022]
Abstract
We have previously demonstrated that PD-1 blockade decreased the incidence of high-grade dysplasia in a carcinogen-induced murine model of oral squamous cell carcinoma (OSCC). It remains unknown, however, whether there are additional factors involved in escape from immune surveillance that could serve as additional targets for immunoprevention. We performed this study to further characterize the immune landscape of oral premalignant lesions (OPL) and determine the impact of targeting of the PD-1, CTLA-4, CD40, or OX40 pathways on the development of OPLs and oral carcinomas in the 4-nitroquinoline 1-oxide model. The immune pathways were targeted using mAbs or, in the case of the PD-1/PD-L1 pathway, using PD-L1-knockout (PD-L1ko) mice. After intervention, tongues and cervical lymph nodes were harvested and analyzed for malignant progression and modulation of the immune milieu, respectively. Targeting of CD40 with an agonist mAb was the most effective treatment to reduce transition of OPLs to OSCC; PD-1 alone or in combination with CTLA-4 inhibition, or PD-L1ko, also reduced progression of OPLs to OSCC, albeit to a lesser extent. Distinct patterns of immune system modulation were observed for the CD40 agonists compared with blockade of the PD-1/PD-L1 axis with or without CTLA-4 blockade; CD40 agonist generated a lasting expansion of experienced/memory cytotoxic T lymphocytes and M1 macrophages, whereas PD-1/CTLA-4 blockade resulted in a pronounced depletion of regulatory T cells among other changes. These data suggest that distinct approaches may be used for targeting different steps in the development of OSCC, and that CD40 agonists merit investigation as potential immunoprevention agents in this setting. PREVENTION RELEVANCE: PD-1/PD-L1 pathway blockade, as well as activation of the CD40 pathway, were able to prevent OPL progression into invasive OSCC in a murine model. A distinct pattern of immune modulation was observed when either the CD40 or the PD-1/PD-L1 pathways were targeted.
Collapse
Affiliation(s)
| | - Taghreed Hirz
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Irene Guijarro
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Monique Nilsson
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marlese A Pisegna
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alissa Poteete
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jared J Fradette
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Limo N Chen
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Don L Gibbons
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiangjun Tian
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey N Myers
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark J McArthur
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Diana Bell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William N William
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Oncology Center, Hospital BP, a Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | - John V Heymach
- Departments of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
31
|
Daub S, Lutgens E, Münzel T, Daiber A. CD40/CD40L and Related Signaling Pathways in Cardiovascular Health and Disease-The Pros and Cons for Cardioprotection. Int J Mol Sci 2020; 21:E8533. [PMID: 33198327 PMCID: PMC7697597 DOI: 10.3390/ijms21228533] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/03/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
The CD40-CD40 ligand (CD40L) dyad represents a scientific and clinical field that has raised many controversies in the past and cannot be clearly defined as being an either beneficial or harmful pathway. Being crucially involved in physiological immunological processes as well as pathological inflammatory reactions, the signaling pathway has been recognized as a key player in the development of both autoimmune and cardiovascular disease. Even though the possibilities of a therapeutic approach to the dyad were recognized decades ago, due to unfortunate events, detailed in this review, pharmacological treatment targeting the dyad, especially in patients suffering from atherosclerosis, is not available. Despite the recent advances in the treatment of classical cardiovascular risk factors, such as arterial hypertension and diabetes mellitus, the treatment of the associated low-grade inflammation that accounts for the progression of atherosclerosis is still challenging. Low-grade inflammation can be detected in a significant portion of patients that suffer from cardiovascular disease and it is therefore imperative to develop new therapeutic strategies in order to combat this driver of atherosclerosis. Of note, established cardiovascular drugs such as angiotensin-converting enzyme inhibitors or statins have proven beneficial cardiovascular effects that are also related to their pleiotropic immunomodulatory properties. In this review, we will discuss the setbacks encountered as well as new avenues discovered on the path to a different, inflammation-centered approach for the treatment of cardiovascular disease with the CD40-CD40L axis as a central therapeutic target.
Collapse
Affiliation(s)
- Steffen Daub
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
| | - Esther Lutgens
- Experimental Vascular Biology Division, Department of Medical Biochemistry, University of Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, 1105 AZ Amsterdam, The Netherlands;
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians Universität, 80336 Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany and Partner Site Munich Heart Alliance, 80336 Munich, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, 55131 Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany; (S.D.); (T.M.)
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, 55131 Mainz, Germany
| |
Collapse
|
32
|
Hübbe ML, Jæhger DE, Andresen TL, Andersen MH. Leveraging Endogenous Dendritic Cells to Enhance the Therapeutic Efficacy of Adoptive T-Cell Therapy and Checkpoint Blockade. Front Immunol 2020; 11:578349. [PMID: 33101304 PMCID: PMC7546347 DOI: 10.3389/fimmu.2020.578349] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 01/15/2023] Open
Abstract
Adoptive cell therapy (ACT), based on treatment with autologous tumor infiltrating lymphocyte (TIL)-derived or genetically modified chimeric antigen receptor (CAR) T cells, has become a potentially curative therapy for subgroups of patients with melanoma and hematological malignancies. To further improve response rates, and to broaden the applicability of ACT to more types of solid malignancies, it is necessary to explore and define strategies that can be used as adjuvant treatments to ACT. Stimulation of endogenous dendritic cells (DCs) alongside ACT can be used to promote epitope spreading and thereby decrease the risk of tumor escape due to target antigen downregulation, which is a common cause of disease relapse in initially responsive ACT treated patients. Addition of checkpoint blockade to ACT and DC stimulation might further enhance response rates by counteracting an eventual inactivation of infused and endogenously primed tumor-reactive T cells. This review will outline and discuss therapeutic strategies that can be utilized to engage endogenous DCs alongside ACT and checkpoint blockade, to strengthen the anti-tumor immune response.
Collapse
Affiliation(s)
- Mie Linder Hübbe
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| | - Ditte Elisabeth Jæhger
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Copenhagen, Denmark
| |
Collapse
|
33
|
Liu B, Shao Y, Liang X, Lu D, Yan L, Churov A, Fu R. CTLA-4 and HLA-DQ are key molecules in the regulation of mDC-mediated cellular immunity by Tregs in severe aplastic anemia. J Clin Lab Anal 2020; 34:e23443. [PMID: 32621335 PMCID: PMC7595925 DOI: 10.1002/jcla.23443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022] Open
Abstract
Background Regulatory T cells (Tregs) inhibit the activation of cluster of differentiation (CD) 4+, CD8+T cells and the antigen‐presenting process of antigen‐presenting cells, and may play an important role in acquired severe aplastic anemia (SAA). Methods Flow cytometry was used to measure CD4+CD25+CD127dim Tregs, cytotoxic T lymphocyte antigen 4 (CTLA‐4) expression on Tregs, and human leukocyte antigen (HLA)‐DQ expression on myeloid dendritic cells (mDCs). The correlations of CTLA‐4 and HLA‐DQ with immune status and clinical indicators and the changes in these indicators after immunosuppressive therapy (IST) were analyzed. Results In SAA patients, the number of Tregs and their CTLA‐4 expression were low but recovered after IST; the HLA‐DQ expression on mDCs was high but decreased after IST. The CTLA‐4 expression on Tregs and the HLA‐DQ expression on mDCs showed a negative correlation. The CTLA‐4 on Tregs was positively but HLA‐DQ on mDCs negatively correlated with the number of Tregs, natural killer (NK) cell number, and CD4+T/CD8+T ratio. CTLA‐4 was positively but HLA‐DQ negatively correlated with the percentage of granulocytoid and erythroid cells in bone marrow, white blood cell count in PB, absolute neutrophil count in PB, and the percentage of reticulocytes in PB. Conclusions CTLA‐4/HLA‐DQ may be key in the regulation of Tregs on mDCs in SAA patients. Our findings should be helpful for further investigation of the mechanism of immune pathogenesis in SAA patients. Studies on the regulators of Treg and CTLA‐4 activity will be valuable for SAA therapeutic target research and disease monitoring.
Collapse
Affiliation(s)
- Bingnan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Yuanyuan Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Xiaowei Liang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Dan Lu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Li Yan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Alexey Churov
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, PR China
| |
Collapse
|
34
|
AbdelMassih AF, AbdelAzeam AS, Ayad A, Kamel AY, Khalil A, Kotb B, Waheed D, Menshawey E, Sefein F, Taha F, Ismail HA, Osman I, Iskander J, El Wakil L, Rashad L, Arsanyous MB, El Shershaby M, Mansour M, Ashraf M, Hafez N, Abuzeid NM, AbdElSalam NMN, Hafez NG, Youssef N, Hozaien R, Saeed R, Kamel D, AbdelHameed MA, Ali S. Unleashing the mysterious link between COVID-19 and a famous childhood vasculitis: Kawasaki disease. EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2020. [PMCID: PMC7358563 DOI: 10.1186/s43054-020-00029-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) emerged as a small outbreak in Wuhan rapidly progressing into the deadliest pandemic since the Spanish flu of 1918. The disease was deemed trivial in children, until the reporting, few days ago, of an emerging pediatric multi-inflammatory syndrome mimicking Kawasaki disease (KD). Main body This report reveals that coronaviridae were implicated in induction of several post-infectious vasculitides, namely, KD, AHEI, and HSP. This occurs in genetically susceptible individuals to vascular inflammation. Shared genetic susceptibilities between KD and CoV include genes encoding for CD 40, HLAB-15:03, and ACE. This leads to augmented inflammation with hypersecretion of cytokines especially IL-6. Conclusion The revealed relationships between KD and CoV can help to predict the risk of KD in COVID-19 patients through screening levels of upregulated cytokines. It might also signify that classic treatment of KD with IVIG might need to be replaced with anti-cytokine therapy in COVID-19 patients.
Collapse
|
35
|
Bosmans LA, Bosch L, Kusters PJH, Lutgens E, Seijkens TTP. The CD40-CD40L Dyad as Immunotherapeutic Target in Cardiovascular Disease. J Cardiovasc Transl Res 2020; 14:13-22. [PMID: 32222950 PMCID: PMC7892683 DOI: 10.1007/s12265-020-09994-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/18/2020] [Indexed: 12/14/2022]
Abstract
Chronic inflammation drives the development of atherosclerosis. Despite optimal treatment of classical cardiovascular risk factors, a substantial portion of the population has elevated inflammatory biomarkers and develops atherosclerosis-related complications, indicating that a residual inflammatory risk drives atherosclerotic cardiovascular disease in these patients. Additional anti-inflammatory therapeutic strategies are therefore required. The co-stimulatory molecule CD40 and its ligand CD40L (CD154) have a central role in the regulation of the inflammatory response during the development of atherosclerosis by modulating the interaction between immune cells and between immune cells and non-immune cells. In this review, we discuss the role of the CD40-CD40L dyad in atherosclerosis, and we discuss recent studies on the therapeutic potential of novel CD40-CD40L targeting strategies in cardiovascular medicine.
Collapse
Affiliation(s)
- Laura A Bosmans
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands
| | - Lena Bosch
- Experimental Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Pascal J H Kusters
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.,Department of Pathology, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University, Munich, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences (ACS), Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 15, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|