1
|
Wu J, Pei F, Zhou L, Li W, Sun R, Li Y, Wang Z, He Z, Zhang X, Jin X, Long Y, Cui W, Wang C, Chen E, Zeng J, Yan J, Lin Q, Zhou F, Huang L, Shang Y, Duan M, Zheng W, Zhu D, Kou Q, Zhang S, Liu Y, Yao C, Shang M, Peng S, Zhou Q, Cheng KK, Guan X. The efficacy and safety of thymosin α1 for sepsis (TESTS): multicentre, double blinded, randomised, placebo controlled, phase 3 trial. BMJ 2025; 388:e082583. [PMID: 39814420 PMCID: PMC11780596 DOI: 10.1136/bmj-2024-082583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE To evaluate whether the immunomodulatory drug thymosin α1 reduces mortality in adults with sepsis. DESIGN Multicentre, double blinded, placebo controlled phase 3 trial. SETTING 22 centres in China, September 2016 to December 2020. PARTICIPANTS 1106 adults aged 18-85 years with a diagnosis of sepsis according to sepsis-3 criteria and randomly assigned in a 1:1 ratio to receive thymosin α1 (n=552) or placebo (n=554). A stratified block method was used for randomisation, and participants were stratified by age (<60 and ≥60 years) and centre. INTERVENTIONS Subcutaneous injection of thymosin α1 or placebo every 12 hours for seven days unless discontinued owing to discharge from the intensive care unit, death, or withdrawal of consent. MAIN OUTCOME MEASURE The primary outcome was 28 day all cause mortality after randomisation. All analyses were based on a modified intention-to-treat set, including participants who received at least one dose of study drug. RESULTS Of 1106 adults with sepsis enrolled in the study, 1089 were included in the modified intention-to-treat analyses (thymosin α1 group n=542, placebo group n=547). 28 day all cause mortality occurred in 127 participants (23.4%) in the thymosin α1 group and 132 (24.1%) in the placebo group (hazard ratio 0.99, 95% confidence interval 0.77 to 1.27; P=0.93 with log-rank test). No secondary or safety outcome differed statistically significantly between the two groups. The prespecified subgroup analysis showed a potential differential effect of thymosin α1 on the primary outcome based on age (<60 years: hazard ratio 1.67, 1.04 to 2.67; ≥60 years: 0.81, 0.61 to 1.09; P for interaction=0.01) and diabetes (diabetes: 0.58, 0.35 to 0.99; no diabetes: 1.16, 0.87 to 1.53; P for interaction=0.04). CONCLUSIONS This trial found no clear evidence to suggest that thymosin α1 decreases 28 day all cause mortality in adults with sepsis. TRIAL REGISTRATION ClinicalTrials.gov NCT02867267.
Collapse
Affiliation(s)
- Jianfeng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
- Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, Guangdong province, China
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Fei Pei
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
- Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, Guangdong province, China
| | - Lixin Zhou
- Department of Critical Care Medicine, The First People's Hospital of Foshan, Foshan, Guangdong province, China
| | - Weiqin Li
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu province, China
| | - Renhua Sun
- Emergency and Critical Care Center, Intensive Care Unit, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang province, China
| | - Yimin Li
- Department of Critical Care Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong province, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, Department of Surgical Intensive Care Unit, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, China
| | - Zhijie He
- Department of Critical Care Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong province, China
| | - Xiaofei Zhang
- Department of Intensive Care Unit, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Xiaodong Jin
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, Sichuan Province, China
| | - Yun Long
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wei Cui
- Department of Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang province, China
| | - Chunting Wang
- Department of Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong province, China
| | - Erzhen Chen
- Department of Emergency, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jun Zeng
- Department of Critical Care Medicine, Guangzhou First People's Hospital, Guangzhou, Guangdong province, China
| | - Jing Yan
- Department of Intensive Care, Zhejiang Hospital, Hangzhou, Zhejiang province, China
| | - Qinhan Lin
- Department of Critical Care Medicine, Affiliated Qingyuan Hospital, The Sixth Clinical Medical School, Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong Province, China
| | - Feihu Zhou
- Department of Critical Care Medicine, The First Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Lei Huang
- Department of Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei province, China
| | - Meili Duan
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Zheng
- Department of Critical Care Medicine, Zhuhai People's Hospital, Zhuhai, Guangdong Province, China
| | - Duming Zhu
- Department of Critical Care Medicine, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qiuye Kou
- Department of Intensive Care Unit, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
- Department of Critical Care Medicine, Foersea Life insurance Guangzhou General Hospital, Guangzhou, Guangdong province, China
| | - Shihong Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Yin Liu
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
| | - Chen Yao
- Peking University First Hospital, Beijing, China
| | - Meixia Shang
- Peking University First Hospital, Beijing, China
| | - Sui Peng
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Qian Zhou
- Clinical Trials Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Kar Keung Cheng
- Department of Applied Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Xiangdong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong province, China
- Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, Guangdong province, China
| |
Collapse
|
2
|
Kologrivova IV, Naryzhnaya NV, Suslova TE. Thymus in Cardiometabolic Impairments and Atherosclerosis: Not a Silent Player? Biomedicines 2024; 12:1408. [PMID: 39061983 PMCID: PMC11273826 DOI: 10.3390/biomedicines12071408] [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: 05/13/2024] [Revised: 06/11/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
The thymus represents a primary organ of the immune system, harboring the generation and maturation of T lymphocytes. Starting from childhood, the thymus undergoes involution, being replaced with adipose tissue, and by an advanced age nearly all the thymus parenchyma is represented by adipocytes. This decline of thymic function is associated with compromised maturation and selection of T lymphocytes, which may directly impact the development of inflammation and induce various autoinflammatory disorders, including atherosclerosis. For a long time, thymus health in adults has been ignored. The process of adipogenesis in thymus and impact of thymic fat on cardiometabolism remains a mysterious process, with many issues being still unresolved. Meanwhile, thymus functional activity has a potential to be regulated, since islets of thymopoeisis remain in adults even at an advanced age. The present review describes the intricate process of thymic adipose involution, focusing on the issues of the thymus' role in the development of atherosclerosis and metabolic health, tightly interconnected with the state of vessels. We also review the recent information on the key molecular pathways and biologically active substances that may be targeted to manipulate both thymic function and atherosclerosis.
Collapse
Affiliation(s)
- Irina V. Kologrivova
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 111A Kievskaya, Tomsk 634012, Russia; (N.V.N.); (T.E.S.)
| | | | | |
Collapse
|
3
|
Garaci E, Paci M, Matteucci C, Costantini C, Puccetti P, Romani L. Phenotypic drug discovery: a case for thymosin alpha-1. Front Med (Lausanne) 2024; 11:1388959. [PMID: 38903817 PMCID: PMC11187271 DOI: 10.3389/fmed.2024.1388959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024] Open
Abstract
Phenotypic drug discovery (PDD) involves screening compounds for their effects on cells, tissues, or whole organisms without necessarily understanding the underlying molecular targets. PDD differs from target-based strategies as it does not require knowledge of a specific drug target or its role in the disease. This approach can lead to the discovery of drugs with unexpected therapeutic effects or applications and allows for the identification of drugs based on their functional effects, rather than through a predefined target-based approach. Ultimately, disease definitions are mostly symptom-based rather than mechanism-based, and the therapeutics should be likewise. In recent years, there has been a renewed interest in PDD due to its potential to address the complexity of human diseases, including the holistic picture of multiple metabolites engaging with multiple targets constituting the central hub of the metabolic host-microbe interactions. Although PDD presents challenges such as hit validation and target deconvolution, significant achievements have been reached in the era of big data. This article explores the experiences of researchers testing the effect of a thymic peptide hormone, thymosin alpha-1, in preclinical and clinical settings and discuss how its therapeutic utility in the precision medicine era can be accommodated within the PDD framework.
Collapse
Affiliation(s)
| | - Maurizio Paci
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
| | - Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigina Romani
- San Raffaele Sulmona, L’Aquila, Italy
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
4
|
Zheng LY, Duan Y, He PY, Wu MY, Wei ST, Du XH, Yao RQ, Yao YM. Dysregulated dendritic cells in sepsis: functional impairment and regulated cell death. Cell Mol Biol Lett 2024; 29:81. [PMID: 38816685 PMCID: PMC11140885 DOI: 10.1186/s11658-024-00602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/21/2024] [Indexed: 06/01/2024] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. Studies have indicated that immune dysfunction plays a central role in the pathogenesis of sepsis. Dendritic cells (DCs) play a crucial role in the emergence of immune dysfunction in sepsis. The major manifestations of DCs in the septic state are abnormal functions and depletion in numbers, which are linked to higher mortality and vulnerability to secondary infections in sepsis. Apoptosis is the most widely studied pathway of number reduction in DCs. In the past few years, there has been a surge in studies focusing on regulated cell death (RCD). This emerging field encompasses various forms of cell death, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death (ADCD). Regulation of DC's RCD can serve as a possible therapeutic focus for the treatment of sepsis. Throughout time, numerous tactics have been devised and effectively implemented to improve abnormal immune response during sepsis progression, including modifying the functions of DCs and inhibiting DC cell death. In this review, we provide an overview of the functional impairment and RCD of DCs in septic states. Also, we highlight recent advances in targeting DCs to regulate host immune response following septic challenge.
Collapse
Affiliation(s)
- Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Peng-Yi He
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Meng-Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Shu-Ting Wei
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xiao-Hui Du
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
- Department of General Surgery, The First Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division of the Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, 100853, China.
| |
Collapse
|
5
|
Kong Y, Chen R, Xu M, Zhang J, Chen G, Hong Z, Zhang H, Dai X, Ma Y, Zhao X, Peng Y, Zhang C, Xing P, Zhang L. Evaluation of the efficacy and safety of a precise thymalfasin-regulated PRaG regimen for advanced refractory solid tumours: protocol for the open-label, prospective, multicentre study (PRaG5.0 study). BMJ Open 2024; 14:e075642. [PMID: 38458816 PMCID: PMC10928769 DOI: 10.1136/bmjopen-2023-075642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 02/07/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION The PRaG regimen, which consists of hypofractionated radiotherapy combined with a programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitor and granulocyte-macrophage colony stimulating factor (GM-CSF), has been demonstrated to have a survival benefit in patients with advanced solid tumours who have failed at least two lines of treatment. Nonetheless, lymphopenia poses an impediment to the enduring efficacy of PD-1/PD-L1 inhibitor therapy. Adequate lymphocyte reserves are essential for the efficacy of immunotherapy. Coupling the PRaG regimen with immunomodulatory agents that augment the number and functionality of lymphocytes may yield further survival benefits in this cohort of patients. OBJECTIVE The aim of this study is to investigate the effectiveness and safety of a meticulously thymalfasin-controlled PRaG regimen in patients with advanced and chemotherapy-resistant solid tumours. METHODS AND ANALYSIS The study has a prospective, single-arm, open-label, multicentre design and aims to recruit up to 60 patients with histologically confirmed advanced solid tumours that have relapsed or metastasised. All eligible patients will receive a minimum of two cycles of the PRaG regimen comprising thymalfasin followed by maintenance treatment with a PD-1/PD-L1 inhibitor and thymalfasin for 1 year or until disease progression. Patients will be monitored according to the predetermined protocol for a year or until disease progression after initiation of radiotherapy. ETHICS AND DISSEMINATION The study protocol was approved by the Ethics Committee of the Second Affiliated Hospital of Soochow University, on 25 November 2022 (JD-LK-2022-151-01) and all other participating hospitals. Findings will be disseminated through national and international conferences. We also plan to publish our findings in high-impact peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT05790447.
Collapse
Affiliation(s)
- Yuehong Kong
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Rongzheng Chen
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Meiling Xu
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Junjun Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Guangqiang Chen
- Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhihui Hong
- Department of Nuclear Medicine, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hong Zhang
- Department of Clinical Laboratory, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaoxiao Dai
- Department of Pathology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yifu Ma
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Xiangrong Zhao
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Yong Peng
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Chenyang Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Pengfei Xing
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, Jiangsu, China
| | - Liyuan Zhang
- Center for Cancer Diagnosis and Treatment, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
6
|
Pei F, Gu B, Miao SM, Guan XD, Wu JF. Clinical practice of sepsis-induced immunosuppression: Current immunotherapy and future options. Chin J Traumatol 2024; 27:63-70. [PMID: 38040590 DOI: 10.1016/j.cjtee.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 12/03/2023] Open
Abstract
Sepsis is a potentially fatal condition characterized by the failure of one or more organs due to a disordered host response to infection. The development of sepsis is closely linked to immune dysfunction. As a result, immunotherapy has gained traction as a promising approach to sepsis treatment, as it holds the potential to reverse immunosuppression and restore immune balance, thereby improving the prognosis of septic patients. However, due to the highly heterogeneous nature of sepsis, it is crucial to carefully select the appropriate patient population for immunotherapy. This review summarizes the current and evolved treatments for sepsis-induced immunosuppression to enhance clinicians' understanding and practical application of immunotherapy in the management of sepsis.
Collapse
Affiliation(s)
- Fei Pei
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Bin Gu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Shu-Min Miao
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Xiang-Dong Guan
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China
| | - Jian-Feng Wu
- Department of Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Clinical Research Center for Critical Care Medicine, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Espinar-Buitrago MS, Vazquez-Alejo E, Magro-Lopez E, Tarancon-Diez L, Leal M, Muñoz-Fernandez MA. Immune modulation via dendritic cells by the effect of Thymosin-alpha-1 on immune synapse in HCMV infection. Int Immunopharmacol 2023; 125:111103. [PMID: 38149577 DOI: 10.1016/j.intimp.2023.111103] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/18/2023] [Accepted: 10/18/2023] [Indexed: 12/28/2023]
Abstract
Tα1 (Thymosin-alpha-1) is a thymus-derived hormone that has been demonstrated to be effective on diverse immune cell subsets. The objective of this study was to determine the in vitro immunomodulatory effect of Tα1 in human cytomegalovirus (HCMV) infection. Dendritic cells (DCs) were isolated from peripheral blood mononuclear cells (PBMCs) by negative selection and cultured in the presence or absence of Tα1. The immunophenotyping of DCs was characterised by multiparametric flow cytometry assessing CD40, CD80, TIM-3 and PDL-1 markers, as well as intracellular TNFα production. Then, autologous CD4+ or CD8+ T-Lymphocytes (TLs) isolated by negative selection from PBMCs were co-cultured with DCs previously treated with Tα1 in the presence or absence of HCMV. Intracellular TNFα, IFNγ, IL-2 production, CD40-L and PD-1 expression were assessed through immunophenotyping, and polyfunctionality in total TLs and memory subsets were evaluated. The results showed that Tα1 increased CD40, CD80, TIM-3 and TNFα intracellular production while decreasing PDL-1 expression, particularly on plasmacytoid dendritic cells (pDCs). Therefore, Tα1 modulated the production of TNFα, IFNγ and IL-2 in both total and memory subsets of CD4+ and CD8+ TLs by upregulating CD40/CD40-L and downregulating PDL-1/PD-1 expression. Our study concludes that Tα1 enhances antigen-presenting capacity of DCs, improves TLs responses to HCMV infection, and enhances the polyfunctionality of CD8+ TLs. Consequently, Tα1 could be an alternative adjuvant for use in therapeutic cell therapy for immunocompromised patients.
Collapse
Affiliation(s)
- M S Espinar-Buitrago
- Sección de Inmunología, Laboratorio Inmuno-Biología Molecular (LIBM), Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanotecnología (CIBER-BBN), Madrid, Spain
| | - E Vazquez-Alejo
- Sección de Inmunología, Laboratorio Inmuno-Biología Molecular (LIBM), Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanotecnología (CIBER-BBN), Madrid, Spain
| | - E Magro-Lopez
- Sección de Inmunología, Laboratorio Inmuno-Biología Molecular (LIBM), Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanotecnología (CIBER-BBN), Madrid, Spain
| | - L Tarancon-Diez
- Sección de Inmunología, Laboratorio Inmuno-Biología Molecular (LIBM), Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanotecnología (CIBER-BBN), Madrid, Spain
| | - M Leal
- Departamento de Medicina Interna, Hospital Viamed Santa Ángela de la Cruz, 41014 Sevilla, Spain; Hogar Residencia de la Santa Caridad, 41001 Sevilla, Spain
| | - M A Muñoz-Fernandez
- Sección de Inmunología, Laboratorio Inmuno-Biología Molecular (LIBM), Hospital General Universitario Gregorio Marañón (HGUGM), Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), 28009 Madrid, Spain; Centro de Investigación Biomédica en Red Bioingeniería, Biomateriales y Nanotecnología (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
8
|
Bai H, Liang L, Qi X, Xu Y, Liu Y, Ren D, Cai Z, Mao W, Wang X, Qin H, Hu F, Shi B. Thymosin α1 modulated the immune landscape of COVID-19 patients revealed by single-cell RNA and TCR sequencing. Int Immunopharmacol 2023; 124:110983. [PMID: 37769533 DOI: 10.1016/j.intimp.2023.110983] [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: 07/12/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND The Coronavirus disease-19 (COVID-19) pandemic has posed a serious threat to global health. Thymosin α1 (Tα1) was considered to be applied in COVID-19 therapy. However, the data remains limited. METHODS Participants with or without Tα1 treatment were recruited. Single cell RNA-sequencing (scRNA-seq) and T cell receptor-sequencing (TCR-seq) of the peripheral blood mononuclear cell (PBMC) samples were done to analyze immune features. The differential expression analysis and functional enrichment analysis were performed to explore the mechanism of Tα1 therapy. RESULTS 33 symptomatic SARS-CoV-2-infected individuals (COV) and 11 healthy controls (HC) were enrolled in this study. The proportion of CD3+ KLRD1+ NKT, TBX21+ CD8+ NKT was observed to increase in COVID-19 patients with Tα1 treatment (COVT) than those without Tα1 (COV) (p = 0.024; p = 0.010). These two clusters were also significantly higher in Health controls with Tα1 treatment (HCT) than those without Tα1 (HC) (p = 0.016; p = 0.031). Besides, a series of genes and pathways related to immune responses were significantly higher enriched in Tα1 groups TBX21+ CD8+ NKT, such as KLRB1, PRF1, natural killer cell-mediated cytotoxicity pathway, chemokine signaling pathway, JAK-STAT signaling pathway. The increased TRBV9-TRBJ1-1 pair existed in both HCs and COVID-19 patients after Tα1 treatment. 1389 common complementarity determining region 3 nucleotides (CDR 3 nt) were found in COV and HC, while 0 CDR 3 nt was common in COVT and HCT. CONCLUSIONS Tα1 increased CD3+ KLRD1+ NKT, TBX21+ CD8+ NKT cell proportion and stimulated the diversity of TCR clones in COVT and HCT. And Tα1 could regulate the expression of genes associated with NKT activation or cytotoxicity to promote NKT cells. These data support the use of Tα1 in COVID-19 patients.
Collapse
Affiliation(s)
- Han Bai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Liyuan Liang
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Xin Qi
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Yao Xu
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Yijia Liu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Doudou Ren
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Zeqiong Cai
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China
| | - Weikang Mao
- LC-BIO TECHNOLOGIES (HANGZHOU) CO., LTD., Hangzhou 310000, China
| | - Xiaorui Wang
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Hongyu Qin
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Fang Hu
- Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China
| | - Bingyin Shi
- The MED-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Building 21, Western China Science and Technology Innovation Harbor, Xi'an 710000, China; Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China.
| |
Collapse
|
9
|
Liu Y, Lu J. Mechanism and clinical application of thymosin in the treatment of lung cancer. Front Immunol 2023; 14:1237978. [PMID: 37701432 PMCID: PMC10493777 DOI: 10.3389/fimmu.2023.1237978] [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: 06/12/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The burden of cancer on public health is becoming more widely acknowledged. Lung cancer has one of the highest incidence and mortality rates of all cancers. The prevalence of early screening, the emergence of targeted therapy, and the development of immunotherapy have all significantly improved the overall prognosis of lung cancer patients. The current state of affairs, however, is not encouraging, and there are issues like poor treatment outcomes for some patients and extremely poor prognoses for those with advanced lung cancer. Because of their potent immunomodulatory capabilities, thymosin drugs are frequently used in the treatment of tumors. The effectiveness of thymosin drugs in the treatment of lung cancer has been demonstrated in numerous studies, which amply demonstrates the potential and future of thymosin drugs for the treatment of lung cancer. The clinical research on thymosin peptide drugs in lung cancer and the basic research on the mechanism of thymosin drugs in anti-lung cancer are both systematically summarized and analyzed in this paper, along with future research directions.
Collapse
Affiliation(s)
| | - Jibin Lu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Tao N, Xu X, Ying Y, Hu S, Sun Q, Lv G, Gao J. Thymosin α1 and Its Role in Viral Infectious Diseases: The Mechanism and Clinical Application. Molecules 2023; 28:molecules28083539. [PMID: 37110771 PMCID: PMC10144173 DOI: 10.3390/molecules28083539] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Thymosin α1 (Tα1) is an immunostimulatory peptide that is commonly used as an immune enhancer in viral infectious diseases such as hepatitis B, hepatitis C, and acquired immune deficiency syndrome (AIDS). Tα1 can influence the functions of immune cells, such as T cells, B cells, macrophages, and natural killer cells, by interacting with various Toll-like receptors (TLRs). Generally, Tα1 can bind to TLR3/4/9 and activate downstream IRF3 and NF-κB signal pathways, thus promoting the proliferation and activation of target immune cells. Moreover, TLR2 and TLR7 are also associated with Tα1. TLR2/NF-κB, TLR2/p38MAPK, or TLR7/MyD88 signaling pathways are activated by Tα1 to promote the production of various cytokines, thereby enhancing the innate and adaptive immune responses. At present, there are many reports on the clinical application and pharmacological research of Tα1, but there is no systematic review to analyze its exact clinical efficacy in these viral infectious diseases via its modulation of immune function. This review offers an overview and discussion of the characteristics of Tα1, its immunomodulatory properties, the molecular mechanisms underlying its therapeutic effects, and its clinical applications in antiviral therapy.
Collapse
Affiliation(s)
- Nana Tao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xie Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yuyuan Ying
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shiyu Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Qingru Sun
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Guiyuan Lv
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jianli Gao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao 999078, China
| |
Collapse
|
11
|
Ke L, Mao W, Shao F, Zhou J, Xu M, Chen T, Liu Y, Tong Z, Windsor J, Ma P, Li W. Association between pretreatment lymphocyte count and efficacy of immune-enhancing therapy in acute necrotising pancreatitis: a post-hoc analysis of the multicentre, randomised, placebo-controlled TRACE trial. EClinicalMedicine 2023; 58:101915. [PMID: 37007743 PMCID: PMC10050769 DOI: 10.1016/j.eclinm.2023.101915] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 04/04/2023] Open
Abstract
Background Immune-enhancing thymosin alpha 1 (Tα1) therapy may reduce infected pancreatic necrosis (IPN) in acute necrotising pancreatitis (ANP). However, the efficacy might be impacted by lymphocyte count due to the pharmacological action of Tα1. In this post-hoc analysis, we tested the hypothesis that pre-treatment absolute lymphocyte count (ALC) determines whether patients with ANP benefit from Tα1 therapy. Methods A post-hoc analysis of data from a multicentre, double-blind, randomised, placebo-controlled trial testing the efficacy of Tα1 therapy in patients with predicted severe ANP was performed. Patients from 16 hospitals of China were randomised to receive a subcutaneous injection of Tα1 1.6 mg every 12 h for the frst 7 days and 1.6 mg once a day for the following 7 days or a matching placebo during the same period. Patients who discontinued the Tα1 regimen prematurely were excluded. Three subgroup analyses were conducted using the baseline ALC (at randomisation), and the group allocation was maintained as intention-to-treat. The primary outcome was the incidence of IPN 90 days after randomisation. The fitted logistic regression model was applied to identify the range of baseline ALC where Tα1 therapy could exert a maximum effect. The original trial is registered with ClinicalTrials.gov, NCT02473406. Findings Between March 18, 2017, and December 10, 2020, a total of 508 patients were randomised in the original trial, and 502 were involved in this analysis, with 248 in the Tα1 group and 254 in the placebo group. Across the three subgroups, there was a uniform trend toward more significant treatment effects in patients with higher baseline ALC. Within the subgroup of patients with baseline ALC≥0.8 × 10ˆ9/L (n = 290), the Tα1 therapy significantly reduced the risk of IPN (covariate adjusted risk difference, -0.12; 95% CI, -0.21,-0.02; p = 0.015). Patients with baseline ALC between 0.79 and 2.00 × 10ˆ9/L benefited most from the Tα1 therapy in reducing IPN (n = 263). Interpretation This post-hoc analysis found that the efficacy of immune-enhancing Tα1 therapy on the incidence of IPN may be associated with pretreatment lymphocyte count in patients with acute necrotising pancreatitis. Funding National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Lu Ke
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, 210010, Jiangsu, China
| | - Wenjian Mao
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
| | - Fang Shao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jing Zhou
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
| | - Minyi Xu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Tao Chen
- Department of Public Health, Policy and Systems, Institute of Population Health, The University of Liverpool, Liverpool, UK
| | - Yuxiu Liu
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, 210010, Jiangsu, China
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Zhihui Tong
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, 210010, Jiangsu, China
- Corresponding author. Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China.
| | - John Windsor
- Surgical and Translational Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Penglin Ma
- Department of Critical Care Medicine, Guiqian International General Hospital, Guiyang, 550004, Guizhou, China
- Corresponding author. Department of Critical Care Medicine, Guiqian International General Hospital, Guiyang, Guizhou Province, 550004, China.
| | - Weiqin Li
- Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, Jiangsu, China
- Department of Critical Care Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, 210010, Jiangsu, China
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, 210010, Jiangsu, China
- Corresponding author. Department of Critical Care Medicine, Jinling Hospital, Medical School of Nanjing University, No. 305 Zhongshan East Road, Nanjing, Jiangsu Province, 210002, China.
| | | |
Collapse
|
12
|
Bellet MM, Renga G, Pariano M, Stincardini C, D'Onofrio F, Goldstein AL, Garaci E, Romani L, Costantini C. COVID-19 and beyond: Reassessing the role of thymosin alpha1 in lung infections. Int Immunopharmacol 2023; 117:109949. [PMID: 36881979 PMCID: PMC9977614 DOI: 10.1016/j.intimp.2023.109949] [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: 12/13/2022] [Revised: 02/15/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023]
Abstract
The recent COVID-19 pandemic has catalyzed the attention of the scientific community to the long-standing issue of lower respiratory tract infections. The myriad of airborne bacterial, viral and fungal agents to which humans are constantly exposed represents a constant threat to susceptible individuals and bears the potential to reach a catastrophic scale when the ease of inter-individual transmission couples with a severe pathogenicity. While we might be past the threat of COVID-19, the risk of future outbreaks of respiratory infections is tangible and argues for a comprehensive assessment of the pathogenic mechanisms shared by airborne pathogens. On this regard, it is clear that the immune system play a major role in dictating the clinical course of the infection. A balanced immune response is required not only to disarm the pathogens, but also to prevent collateral tissue damage, thus moving at the interface between resistance to infection and tolerance. Thymosin alpha1 (Tα1), an endogenous thymic peptide, is increasingly being recognized for its ability to work as an immunoregulatory molecule able to balance a derailed immune response, working as immune stimulatory or immune suppressive in a context-dependent manner. In this review, we will take advantage from the recent work on the COVID-19 pandemic to reassess the role of Tα1 as a potential therapeutic molecule in lung infections caused by either defective or exaggerated immune responses. The elucidation of the immune regulatory mechanisms of Tα1 might open a new window of opportunity for the clinical translation of this enigmatic molecule and a potential new weapon in our arsenal against lung infections.
Collapse
Affiliation(s)
- Marina M Bellet
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giorgia Renga
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Fiorella D'Onofrio
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Enrico Garaci
- MEBIC Consortium and University San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| |
Collapse
|
13
|
Nevo N, Lee Goldstein A, Bar-David S, Abu-Abeid A, Dayan D, Lahat G, Nizri E. Immunological effects of heated intraperitoneal chemotherapy can be augmented by thymosin α1. Int Immunopharmacol 2023; 116:109829. [PMID: 36758296 DOI: 10.1016/j.intimp.2023.109829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Peritoneal metastases of colorectal carcinoma origin (PM-CRC) are treated by cytoreductive surgery and heated intraperitoneal chemotherapy (HIPEC). However, the majority of patients recur, calling for novel treatments. We explored the immunogenic changes induced by HIPEC and the possibility to use thymosin α1 (Tα1) as an immune-stimulatory agent. METHODS We used an experimental murine model of PM-CRC combined with mitomycin (MMC)-based HIPEC. We determined immune cell infiltration into tumor metastases after HIPEC administration by means of immunohistochemistry, and determined immunogenic cell death signals in tumor cells by real-time polymerase chain reaction. RESULTS Mice with PM-CRC treated by HIPEC had increased overall survival (OS) compared to sham-treated mice (median OS 22.8 vs 18.9 days, respectively; P < 0.001). HIPEC induced increased infiltration of CD4+, CD8+, CD68 + and CD20 + cells into omental and visceral metastases at a magnitude of 40-100 %. We searched for potential immune signals induced by HIPEC by determining its effects on known immunogenic cell death proteins (heat-shock protein [HSP]-70, HSP-90 and calreticulin). HIPEC significantly increased HSP-90 mRNA expression (2.37 ± 1.5 vs 1-fold change, P < 0.05). The OS of Tα1 treated mice significantly improved compared to HIPEC-treated mice (16.3 ± 0.8 vs 14.1 ± 0.6 days, respectively, P = 0.02) and vs sham (11.8 ± 0.8 days, P = 0.007). CONCLUSIONS HIPEC induced immunogenic changes that led to increased immune cell infiltration. These changes were further augmented by Tα1 treatment. Future studies aimed at optimizing Tα1 treatment should focus upon the immune response it evokes.
Collapse
Affiliation(s)
- Nadav Nevo
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | | | - Shoshi Bar-David
- Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Adam Abu-Abeid
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel
| | - Danit Dayan
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel
| | - Guy Lahat
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Nizri
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
14
|
Thymosin α-1 in cancer therapy: Immunoregulation and potential applications. Int Immunopharmacol 2023; 117:109744. [PMID: 36812669 DOI: 10.1016/j.intimp.2023.109744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 02/22/2023]
Abstract
Thymosin α-1 (Tα-1) is an immunomodulating polypeptide of 28 amino acids, which was the first peptide isolated from thymic tissue and has been widely used for the treatment of viral infections, immunodeficiencies, and especially malignancies. Tα-1 stimulates both innate and adaptive immune responses, and its regulation of innate immune cells and adaptive immune cells varies under different disease conditions. Pleiotropic regulation of immune cells by Tα-1 depends on activation of Toll-like receptors and its downstream signaling pathways in various immune microenvironments. For treatment of malignancies, the combination of Tα-1 and chemotherapy has a strong synergistic effect by enhancing the anti-tumor immune response. On the basis of the pleiotropic effect of Tα-1 on immune cells and the promising results of preclinical studies, Tα-1 may be a favorable immunomodulator to enhance the curative effect and decrease immune-related adverse events of immune checkpoint inhibitors to develop novel cancer therapies.
Collapse
|
15
|
Zhang D, Wang L, Wang Z, Shi X, Tang W, Jiang L, Bo Ran Yi BYCH, Lv X, Hu C, Xiao D. Immunological responses of septic rats to combination therapy with thymosin α1 and vitamin C. Open Life Sci 2023; 18:20220551. [PMID: 36816800 PMCID: PMC9922062 DOI: 10.1515/biol-2022-0551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 02/10/2023] Open
Abstract
This study investigated the effect of combined thymosin α1 and vitamin C (Tα1 + VitC) on the immunological responses of septic rats. Five groups were designed. The septic model was established by the cecal ligation puncture (CLP) method. The sham group did not undergo CLP, the model group was given normal saline solution, the Tα1 group was given Tα1 (200 µg/kg), the VitC group was given VitC (200 mg/kg), and the Tα1 + VitC group was given Tα1 + VitC. Specimens for immunological analyses were collected at 6, 12, 24, and 48 h posttreatment in each group except for the sham group (only at 48 h). CD4 + CD25 + T cells in the peripheral blood and dendritic cell (DC) proportions in the spleen were analyzed by flow cytometry. Tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), transforming growth factor-β (TGF-ß1), and nuclear factor kappa-B (NF-κB) were measured by ELISA. CD4 + CD25 + T cells and OX62 + DCs levels significantly increased in the model group and decreased in the Tα1 and/or VitC treatment groups. Similarly, the levels of TNF-α, IL-6, TGF-ß1, and NF-κB significantly increased in the model group and decreased in the Tα1, VitC, and Tα1 + VitC groups, indicating that combined Tα1 and VitC therapy may help regulate the immunological state of patients with sepsis, thereby improving prognosis.
Collapse
Affiliation(s)
- Daquan Zhang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Lu Wang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Zhigao Wang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Xiaohui Shi
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Wen Tang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Long Jiang
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Ba Yin Cha Han Bo Ran Yi
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Xinwei Lv
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Congyu Hu
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| | - Dong Xiao
- Department of Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi 830001, Xinjiang, China
| |
Collapse
|
16
|
Bencze D, Fekete T, Pfliegler W, Szöőr Á, Csoma E, Szántó A, Tarr T, Bácsi A, Kemény L, Veréb Z, Pázmándi K. Interactions between the NLRP3-Dependent IL-1β and the Type I Interferon Pathways in Human Plasmacytoid Dendritic Cells. Int J Mol Sci 2022; 23:12154. [PMID: 36293012 PMCID: PMC9602791 DOI: 10.3390/ijms232012154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Generally, a reciprocal antagonistic interaction exists between the antiviral type I interferon (IFN) and the antibacterial nucleotide-binding oligomerization domain (NOD)-like receptor pyrin domain containing 3 (NLRP3)-dependent IL-1β pathways that can significantly shape immune responses. Plasmacytoid dendritic cells (pDCs), as professional type I IFN-producing cells, are the major coordinators of antiviral immunity; however, their NLRP3-dependent IL-1β secretory pathway is poorly studied. Our aim was to determine the functional activity of the IL-1β pathway and its possible interaction with the type I IFN pathway in pDCs. We found that potent nuclear factor-kappa B (NF-κB) inducers promote higher levels of pro-IL-1β during priming compared to those activation signals, which mainly trigger interferon regulatory factor (IRF)-mediated type I IFN production. The generation of cleaved IL-1β requires certain secondary signals in pDCs and IFN-α or type I IFN-inducing viruses inhibit IL-1β production of pDCs, presumably by promoting the expression of various NLRP3 pathway inhibitors. In line with that, we detected significantly lower IL-1β production in pDCs of psoriasis patients with elevated IFN-α levels. Collectively, our results show that the NLRP3-dependent IL-1β secretory pathway is inducible in pDCs; however, it may only prevail under inflammatory conditions, in which the type I IFN pathway is not dominant.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Walter Pfliegler
- Department of Molecular Biotechnology and Microbiology, Faculty of Science and Technology, University of Debrecen, 4032 Debrecen, Hungary
| | - Árpád Szöőr
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Eszter Csoma
- Department of Medical Microbiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Antónia Szántó
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tünde Tarr
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Attila Bácsi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Lajos Kemény
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Zoltán Veréb
- Regenerative Medicine and Cellular Pharmacology Laboratory, Department of Dermatology and Allergology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Chu C, Liang Y, Lin X, Liu Y, Liu S, Guo J, Wang D, Wang J, Liu H, Qiu B. Hypofractionated Radiation Therapy Combined With Weekly Chemotherapy in Patients With Unresectable or Recurrent Thymic Epithelial Tumor: A Prospective, Single-Arm Phase 2 Study (GASTO-1042). Int J Radiat Oncol Biol Phys 2022; 114:89-98. [PMID: 35598797 DOI: 10.1016/j.ijrobp.2022.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 04/16/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE This prospective phase 2 study aimed to evaluate the efficacy and safety of hypofractionated radiation therapy (HRT) combined with concurrent weekly chemotherapy in patients with unresectable or recurrent thymic epithelial tumors (TETs). METHODS AND MATERIALS Patients with unresectable or recurrent intrathoracic TETs that could be encompassed within the radiation fields were enrolled. HRT using intensity modulated radiation therapy (IMRT) technique was administered with 3 different levels of radiation doses (51 Gy/17 fractions (fx), 48 Gy/12 fx, and 45 Gy/9 fx; biologically effective dose of 66.3-67.5Gy), combined with weekly docetaxel (25 mg/m2) and nedaplatin (25 mg/m2). Weekly thymosin α1 (1.6 mg) was administered from the start to 2 months after radiation therapy. The objective response rate (ORR), progression-free survival (PFS), overall survival (OS), health-related quality of life (QOL), and toxicity were recorded. RESULTS Fifty eligible patients enrolled from August 1, 2018, to July 1, 2020, were analyzed. Most patients (82.0%) had stage IVB tumors. Patients had IMRT-HRT (36-51 Gy in 9-17 fx, median biologically effective dose of 67.2 Gy) and concurrent weekly docetaxel/nedaplatin (2-4 cycles). During a median follow-up of 25.0 months (14.0-40.0), the ORR was 83.7%, the 2-year PFS was 59.1%, and the 2-year OS was 90.0%. There was 1 (2.0%) in-field recurrence while 19 (38.0%) patients developed out-of-field recurrence. Grade 3 pneumonitis was observed in 1 patient (2.0%). The ORR, 2-year PFS, 2-year OS, and toxicity were similar among 3 dose levels. Fourteen (28.0%) patients had 2 to 4 courses of radiation therapy because of recurrent diseases. Only 1 suffered from grade 1 pulmonary fibrosis during follow-up. Most patients (88%) maintained a stable QOL within 1 year after radiation therapy. CONCLUSIONS IMRT-HRT and concurrent weekly docetaxel/nedaplatin was effective and well tolerated in unresectable or recurrent TETs. Considering the common out-of-field recurrence, this combined regimen could be an option for repeated radiation therapy. Thymosin α1 might help lower the incidence of pneumonitis and maintain the QOL.
Collapse
Affiliation(s)
- Chu Chu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Ying Liang
- State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China; Departments of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiaosheng Lin
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Yimei Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Songran Liu
- State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China; Departments of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinyu Guo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Daquan Wang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China
| | - Junye Wang
- State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China; Departments of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China.
| | - Bo Qiu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China; State Key Laboratory of Oncology in South China, Guangzhou, China; Collaborative Innovation Center for Cancer Medicine; Lung Cancer Institute, Sun Yat-sen University, Guangzhou, China; Guangdong Association Study of Thoracic Oncology, Guangzhou, China.
| |
Collapse
|
18
|
Thymosin alpha 1 as an adjuvant to hyperthermic intraperitoneal chemotherapy in an experimental model of peritoneal metastases from colonic carcinoma. Int Immunopharmacol 2022; 111:109166. [PMID: 35994852 DOI: 10.1016/j.intimp.2022.109166] [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: 05/13/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Heated intraperitoneal chemotherapy (HIPEC) is currently implemented in the treatment of peritoneal metastases from colorectal carcinoma (PM-CRC) origin. However, recurrence is common and the effectiveness of HIPEC has been questioned. The aim of this study was to evaluate the use of thymosin alpha 1 (Tα1), an immunomodulatory molecule, as an adjuvant to HIPEC treatment. METHODS We developed an experimental model of HIPEC by the induction of PM-CRC in C57BL mice and intra-abdominal perfusion of mitomycin C (MMC). Mice were treated with Tα1 at 0.6 mg/kg for 5 days after HIPEC. Clinical and immunological parameters were compared between HIPEC and HIPEC + Tα1 groups. RESULTS Treatment with Tα1 increased overall survival of mice compared to HIPEC treatment alone and sham-treated animals (16.1 ± 0.8 vs. 14.1 ± 0.6 and 11.8 ± 0.8, respectively, p = 0.02). Tα1 had no direct anti-tumor effect, as seen by lack of inhibition of tumor cell proliferation. Tα1 treatment induced a T helper (Th) 1 immune response in tumor metastases as evidenced by a significant increase of the Th1-specific markers IFN-γ and T-bet (1.21 ± 0.3 vs. 0.52 ± 0.08, p < 0.05; 0.88 ± 0.04 vs. 0.64 ± 0.14, p < 0.05, respectively). This Th1 skew was accompanied by increased CD8+ infiltration into omental and visceral metastases by Tα1 treatment compared to sham and HIPEC-treated animals (21.24 ± 2.16 vs. 10.45 ± 0.89 and 7.7 ± 1.3, p < 0.001; 14.12 ± 1.54 vs. 12.12 ± 0.01 and 6.64 ± 0.87, p < 0.01, respectively). CONCLUSIONS Tα1 augments the effect of HIPEC by the induction of a Th1 anti-tumor immune response. Further experiments should evaluate Tα1 and other novel immunomodulators in order to exploit the immunological opportunities created by HIPEC.
Collapse
|
19
|
Liu F, Qiu B, Xi Y, Luo Y, Luo Q, Wu Y, Chen N, Zhou R, Guo J, Wu Q, Xiong M, Liu H. Efficacy of thymosin α1 in management of radiation pneumonitis in patients with locally advanced non-small cell lung cancer treated with concurrent chemoradiotherapy: A Phase 2 Clinical Trial (GASTO-1043). Int J Radiat Oncol Biol Phys 2022; 114:433-443. [PMID: 35870709 DOI: 10.1016/j.ijrobp.2022.07.009] [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: 02/07/2022] [Revised: 06/26/2022] [Accepted: 07/03/2022] [Indexed: 10/31/2022]
Abstract
PURPOSE To evaluate the efficacy of thymosin α1 in management of radiation pneumonitis (RP) in patients with locally advanced non-small cell lung cancer (LANSCLC) treated with concurrent chemoradiotherapy (CCRT). METHODS AND MATERIALS This phase II, single-arm trial enrolled patients with unresectable LANSCLC of 18 to 75 years' old and an Eastern Cooperative Oncology Group performance status of 0 to 1. Eligible patients received definitive CCRT and weekly thymosin α1 from the start of CCRT till 2 months after CCRT. Patients were administered 51 Gy in 17 daily fractions or 40 Gy in 10 daily fractions in the first course followed by a re-evaluation and those patients without disease progression had an adaptive plan of 15 Gy in 5 daily fractions or 24 Gy in 6 daily fractions as a boost. Concurrent chemotherapy consisted of weekly docetaxel (25 mg/m2) and nedaplatin (25 mg/m2) during radiation therapy. The primary endpoint was the incidence of Grade (G) ≥2 RP. Secondary endpoints included the incidence of late pulmonary fibrosis, total lymphocyte count (TLC), serum C-reactive protein (CRP) levels, and the composition of gut microbiota. TLC and CRP data were collected at baseline, 2-3 weeks during CCRT, the end of CCRT, 2 and 6 months after CCRT. Fecal samples were collected at baseline and the end of CCRT. Patients treated with CCRT but without thymosin α1 intervention during the same period were selected as the control group by the propensity score matching method. RESULTS Sixty-nine patients were enrolled in the study, and another 69 patients were selected as the control group. The incidence of G≥2 RP was lower in the study group compared with control cases (36.2% vs 53.6%, P=0.040). G1 late pulmonary fibrosis occurred in 2 (3.7%) patients of the control group compared with no event in the study group (P=0.243). Compared with the control group, the incidence of G3-4 lymphopenia (19.1% vs. 62.1%, P<0.001) was lower, and the median TLC nadir (0.51 k/µL vs. 0.30 k/µL, P<0.001) was higher in the study group. The proportion of patients with maximum CRP≥100 mg/L was lower in the study group (13.8% vs. 29.7% P=0.029). The diversity and community composition of the gut microbiota were not significantly different between the two groups. CONCLUSIONS Administration of thymosin α1 during and after CCRT was associated with significant reductions in G≥2 RP and G3-4 lymphopenia in patients with LANSCLC compared to historic controls.
Collapse
Affiliation(s)
- Fangjie Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Bo Qiu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Yu Xi
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China; School of biology and biological engineering, South China University of Technology, Guangzhou. China
| | - Yifeng Luo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou
| | - Qiaoting Luo
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Yingjia Wu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Naibin Chen
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Rui Zhou
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Jinyu Guo
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Mai Xiong
- Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou
| | - Hui Liu
- Department of Radiation Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‑sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
20
|
Changes in immune function and immunomodulatory treatments of septic patients. Clin Immunol 2022; 239:109040. [DOI: 10.1016/j.clim.2022.109040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/23/2022] [Accepted: 05/07/2022] [Indexed: 12/25/2022]
|
21
|
Badr El-Din NK, Othman AI, Amer ME, Ghoneum M. Thymax, a gross thymic extract, exerts cell cycle arrest and apoptosis in Ehrlich ascites carcinoma in vivo. Heliyon 2022; 8:e09047. [PMID: 35299600 PMCID: PMC8920936 DOI: 10.1016/j.heliyon.2022.e09047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/01/2021] [Accepted: 03/01/2022] [Indexed: 11/30/2022] Open
Abstract
Thymax is a gross thymic extract that has been shown to induce apoptosis in vitro for human breast cancer cells. Here we examine Thymax's ability to induce apoptosis in animals bearing Ehrlich ascites carcinoma (EAC). Thymax was administered six days/week orally to mice (5.45 mg/kg body weight) beginning either 14 days prior to EAC inoculation or 9 days post inoculation; treatment continued for 30 days post inoculation. Pretreatment of mice with Thymax markedly delayed tumor growth and reduced tumor incidence by 38.9%, and tumor volumes relative to untreated controls were suppressed by 90.5% and 55.0% for pre- and post-inoculation groups, respectively. Treatment with Thymax inhibited cellular proliferation by decreasing the expression of tumor markers Ki-67, PCNA, and Cyclin D1 in cancer cells and increasing the expression of p21 and p27. This was associated with the ability of Thymax to arrest the cell cycle of EAC cells in the G0/G1 phase and to induce apoptosis, as indicated by a significant increase in the sub-G1 phase's percentage of hypodiploid cells and further affirmed by DNA fragmentation and Annexin V/propidium iodide staining. In addition, Thymax exerted its apoptotic effect in EAC cancer cells through a mitochondrial-dependent pathway, as evidenced by an increased Bax/Bcl-2 ratio, up-regulation of p53 expression, and activation of caspase-3. We conclude that Thymax supplementation enhances tumor cell demise by arresting the cell cycle and inducing apoptosis. These data suggest that Thymax could be a new adjuvant for breast cancer treatment.
Collapse
Affiliation(s)
- Nariman K. Badr El-Din
- Department of Zoology, Faculty of Science, University of Mansoura, Mansoura 33516, Egypt
| | - Azza I. Othman
- Department of Zoology, Faculty of Science, University of Mansoura, Mansoura 33516, Egypt
| | - Maggie E. Amer
- Department of Zoology, Faculty of Science, University of Mansoura, Mansoura 33516, Egypt
| | - Mamdooh Ghoneum
- Department of Surgery, Charles R. Drew University of Medicine and Science, Los Angeles, CA, USA
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
22
|
Al-Suhaimi EA, Aljafary MA, Alkhulaifi FM, Aldossary HA, Alshammari T, AL-Qaaneh A, Aldahhan R, Alkhalifah Z, Gaymalov ZZ, Shehzad A, Homeida AM. Thymus Gland: A Double Edge Sword for Coronaviruses. Vaccines (Basel) 2021; 9:1119. [PMID: 34696231 PMCID: PMC8539924 DOI: 10.3390/vaccines9101119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 02/06/2023] Open
Abstract
The thymus is the main lymphoid organ that regulates the immune and endocrine systems by controlling thymic cell proliferation and differentiation. The gland is a primary lymphoid organ responsible for generating mature T cells into CD4+ or CD8+ single-positive (SP) T cells, contributing to cellular immunity. Regarding humoral immunity, the thymic plasma cells almost exclusively secrete IgG1 and IgG3, the two main complement-fixing effector IgG subclasses. Deformity in the thymus can lead to inflammatory diseases. Hassall's corpuscles' epithelial lining produces thymic stromal lymphopoietin, which induces differentiation of CDs thymocytes into regulatory T cells within the thymus medulla. Thymic B lymphocytes produce immunoglobulins and immunoregulating hormones, including thymosin. Modulation in T cell and naive T cells decrement due to thymus deformity induce alteration in the secretion of various inflammatory factors, resulting in multiple diseases. Influenza virus activates thymic CD4+ CD8+ thymocytes and a large amount of IFNγ. IFNs limit virus spread, enhance macrophages' phagocytosis, and promote the natural killer cell restriction activity against infected cells. Th2 lymphocytes-produced cytokine IL-4 can bind to antiviral INFγ, decreasing the cell susceptibility and downregulating viral receptors. COVID-19 epitopes (S, M, and N proteins) with ≥90% identity to the SARS-CoV sequence have been predicted. These epitopes trigger immunity for antibodies production. Boosting the immune system by improving thymus function can be a therapeutic strategy for preventing virus-related diseases. This review aims to summarize the endocrine-immunoregulatory functions of the thymus and the underlying mechanisms in the prevention of COVID-19.
Collapse
Affiliation(s)
- Ebtesam A. Al-Suhaimi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Meneerah A. Aljafary
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Fadwa M. Alkhulaifi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| | - Hanan A. Aldossary
- Epidemic Diseases Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; or
| | - Thamer Alshammari
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
| | - Ayman AL-Qaaneh
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
- Clinical Pharmacy Services Division, Pharmacy Services Department, Johns Hopkins Aramco Healthcare (JHAH), Dhahran 31311, Saudi Arabia
| | - Razan Aldahhan
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Zahra Alkhalifah
- Genetic Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (T.A.); (A.A.-Q.); (Z.A.)
| | - Zagit Z. Gaymalov
- Earlystage OÜ, Lasnamäe Linnaosa, Sepapaja tn 6, Harju Maakond, 15551 Tallinn, Estonia;
| | - Adeeb Shehzad
- Clinical Pharmacy Research Department, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia;
| | - Abdelgadir M. Homeida
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; (M.A.A.); (F.M.A.); (A.M.H.)
| |
Collapse
|
23
|
Mehta Y, Dixit SB, Zirpe K, Sud R, Gopal PB, Koul PA, Mishra VK, Ansari AS, Chamle VS. Therapeutic Approaches in Modulating the Inflammatory and Immunological Response in Patients With Sepsis, Acute Respiratory Distress Syndrome, and Pancreatitis: An Expert Opinion Review. Cureus 2021; 13:e18393. [PMID: 34692364 PMCID: PMC8526068 DOI: 10.7759/cureus.18393] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2021] [Indexed: 12/15/2022] Open
Abstract
Immunomodulation has long been an adjunct approach in treating critically ill patients with sepsis, acute respiratory distress syndrome (ARDS), and acute pancreatitis (AP). Hyperactive immune response with immunopathogenesis leads to organ dysfunction and alters the clinical outcomes in critically ill. Though the immune response in the critically ill might have been overlooked, it has gathered greater attention during this novel coronavirus disease 2019 (COVID-19) pandemic. Modulating hyperactive immune response, the cytokine storm, especially with steroids, has shown to improve the outcomes in COVID-19 patients. In this review, we find that immune response pathogenesis in critically ill patients with sepsis, ARDS, and AP is nearly similar. The use of immunomodulators such as steroids, broad-spectrum serine protease inhibitors such as ulinastatin, thymosin alpha, intravenous immunoglobulins, and therapies such as CytoSorb and therapeutic plasma exchange may help in improving the clinical outcomes in these conditions. As the experience of the majority of physicians in using such therapeutics may be limited, we provide our expert comments regarding immunomodulation to optimize outcomes in patients with sepsis/septic shock, ARDS, and AP.
Collapse
Affiliation(s)
- Yatin Mehta
- Institute of Critical Care and Anesthesiology, Medanta - The Medicity, Gurugram, IND
| | | | - Kapil Zirpe
- Neurocritical Care, Grant Medical Foundation, Ruby Hall Clinic, Pune, IND
| | - Randhir Sud
- Institute of Digestive & Hepatobiliary Sciences, Medanta - The Medicity, Gurugram, IND
| | - Palepu B Gopal
- Department of Critical Care, Continental Hospitals, Hyderabad, IND
| | - Parvaiz A Koul
- Department of Pulmonary Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, IND
| | - Vijay K Mishra
- Medica Institute of Critical Care, Bhagwan Mahavir Medica Superspecialty Hospital, Ranchi, IND
| | - Abdul S Ansari
- Department of Critical Care Services, Nanavati Super Specialty Hospital, Mumbai, IND
| | | |
Collapse
|
24
|
Case NT, Duah K, Larsen B, Wong CJ, Gingras AC, O'Meara TR, Robbins N, Veri AO, Whitesell L, Cowen LE. The macrophage-derived protein PTMA induces filamentation of the human fungal pathogen Candida albicans. Cell Rep 2021; 36:109584. [PMID: 34433036 PMCID: PMC8454912 DOI: 10.1016/j.celrep.2021.109584] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/23/2021] [Accepted: 07/29/2021] [Indexed: 12/01/2022] Open
Abstract
Evasion of killing by immune cells is crucial for fungal survival in the host. For the human fungal pathogen Candida albicans, internalization by macrophages induces a transition from yeast to filaments that promotes macrophage death and fungal escape. Nutrient deprivation, alkaline pH, and oxidative stress have been implicated as triggers of intraphagosomal filamentation; however, the impact of other host-derived factors remained unknown. Here, we show that lysates prepared from macrophage-like cell lines and primary macrophages robustly induce C. albicans filamentation. Enzymatic treatment of lysate implicates a phosphorylated protein, and bioactivity-guided fractionation coupled to mass spectrometry identifies the immunomodulatory phosphoprotein PTMA as a candidate trigger of C. albicans filamentation. Immunoneutralization of PTMA within lysate abolishes its activity, strongly supporting PTMA as a filament-inducing component of macrophage lysate. Adding to the known repertoire of physical factors, this work implicates a host protein in the induction of C. albicans filamentation within immune cells. The human fungal pathogen Candida albicans filaments within host macrophages, enabling its escape. Case et al. demonstrate that lysates prepared from macrophage-like cell lines and primary macrophages induce C. albicans filamentation and implicate the immunomodulatory protein prothymosin alpha (PTMA) as a trigger of filamentation produced by host immune cells.
Collapse
Affiliation(s)
- Nicola T Case
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Kwamaa Duah
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Brett Larsen
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Cassandra J Wong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Teresa R O'Meara
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Amanda O Veri
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Luke Whitesell
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Leah E Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
25
|
Li HP, He X, Zhang L, Li CX, Li SQ, Li QY. Therapeutic Agents Rounding Up the Immunopathology of COVID-19. Ther Clin Risk Manag 2021; 17:657-668. [PMID: 34234442 PMCID: PMC8254585 DOI: 10.2147/tcrm.s313003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 pandemic has caused more than 3 million deaths globally during the past year. The direct attack from SARS-CoV-2 and hyperactivated immune response contribute to the progress and deterioration of COVID-19. After the virus invades, the activation and release of cytokines/chemokines cause "cytokine storm", leading to acute respiratory distress syndrome (ARDS) and multiple organs dysfunction syndrome (MODS). Eliminating virus and blocking cytokines are important checkpoints of COVID-19 therapy, and several agents targeting immunopathology, including interferons, thymosin, glucocorticoids and immunoglobulin, have shown therapeutic effects in severe patients with COVID-19. Herein, we reviewed the practice evidences and concluded that several agents rounding up the immunopathology of COVID-19 may be the alternative approaches under the scenario of the lacking of effective antiviral drugs.
Collapse
Affiliation(s)
- Hong Peng Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Xuan He
- Department of Pharmacy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Liu Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Chuan Xiang Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Department of Respiratory Medicine, Wuhan No.3 Hospital, Wuhan, 430000, People’s Republic of China
| | - Shi Qi Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
- Institute of Respiratory Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People’s Republic of China
| |
Collapse
|
26
|
Zheng QN, Xu MY, Gan FM, Ye SS, Zhao H. Thymosin as a possible therapeutic drug for COVID-19: A case report. World J Clin Cases 2021; 9:4090-4094. [PMID: 34141770 PMCID: PMC8180217 DOI: 10.12998/wjcc.v9.i16.4090] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND There are no effective antiviral therapies for coronavirus disease 2019 (COVID-19) at present. Although most patients with COVID-19 have a mild or moderate course of disease, up to 5%-10% of patients may have a serious and potentially life-threatening condition, indicating an urgent need for effective therapeutic drugs. The therapeutic effect of thymosin on COVID-19 has not been previously studied. In this paper, for the first time we report a case of thymosin treatment of COVID-19.
CASE SUMMARY A 51-year-old man with imported COVID-19 was admitted with definite symptoms of chest tightness, chest pain, and fatigue. The polymerase chain reaction results for severe acute respiratory syndrome coronavirus 2 were negative. The antibody test was positive, confirming the diagnosis of COVID-19. As many orally administered drugs were not well tolerated due to gastrointestinal symptoms, an emergency use of thymosin, a polypeptide consisting of 28 amino acids, was administered by injection. Finally, after the implementation of the treatment program, symptoms and lung imaging improved significantly.
CONCLUSION In this case report, it is confirmed that thymosin may help alleviate the severity of COVID-19 symptoms.
Collapse
Affiliation(s)
- Qiong Na Zheng
- Department of Infectious Diseases, The Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, Zhejiang Province, China
| | - Mei Yan Xu
- Department of Infectious Diseases, The Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, Zhejiang Province, China
| | - Fang Min Gan
- Department of Infectious Diseases, The Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, Zhejiang Province, China
| | - Sha Sha Ye
- Department of Infectious Diseases, The Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, Zhejiang Province, China
| | - Hui Zhao
- Department of Infectious Diseases, The Affiliated Yueqing Hospital, Wenzhou Medical University, Yueqing 325600, Zhejiang Province, China
| |
Collapse
|
27
|
Feng Y, Zhu G, Lang S, Hao P, Li G, Chen F, Zhuo W, Duan Y, Zhang A, Chen Z, Sun J. The Efficacy and Safety of Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitor Combined With Thymosin in Advanced Non-Small Cell Lung Cancer Patients Harboring Active Epidermal Growth Factor Receptor Mutations. Front Oncol 2021; 11:659065. [PMID: 34123814 PMCID: PMC8195272 DOI: 10.3389/fonc.2021.659065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/03/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To explore the efficacy and safety of EGFR-TKI combined with thymosin therapy in advanced non-small cell lung cancer (NSCLC) patients harboring active EGFR mutations. Methods Patients confirmed as advanced NSCLC with active EGFR mutations were recruited from August 2008 to July 2018 retrospectively. Patients treated with EGFR-TKI were classified as the EGFR-TKI group. And those received EGFR-TKI and thymosin therapy were designated as the EGFR-TKI plus thymosin group. The primary endpoint was progression-free survival (PFS). The secondary endpoints included overall survival (OS), tumor response and adverse effects. Results The median PFS was significantly longer in EGFR-TKI plus thymosin group than that in EGFR-TKI group (14.4 months vs. 9.2 months; HR=0.433, 95% CI 0.322 - 0.582, P<0.0001). The median OS was also prolonged in EGFR-TKI plus thymosin group than that in EGFR-TKI group (29.5 months vs. 19.8 months; HR=0.430, 95% CI 0.319 - 0.580, P<0.0001). The objective response rate in EGFR-TKI plus thymosin group and EGFR-TKI group were 60.0% versus 60.8% (P=0.918). The disease control rate was 96.9% in EGFR-TKI plus thymosin group and 97.7% in EGFR-TKI group (P=1.000). There were no significant differences in adverse effects between the two groups. The number of CD3+T cells in peripheral blood decreased significantly after treatment including both CD3+CD4+T and CD3+CD8+T subsets in EGFR-TKI group, but not in EGFR-TKI plus thymosin group. Conclusions Combination of EGFR-TKI and thymosin can significantly prolong the PFS and OS compared with EGFR-TKI monotherapy without more adverse events, which offers a new strategy in clinic.
Collapse
Affiliation(s)
- Yongdong Feng
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Guangkuo Zhu
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Song Lang
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Ping Hao
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Guanghui Li
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Fanglin Chen
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wenlei Zhuo
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yuzhong Duan
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Anmei Zhang
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhengtang Chen
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jianguo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
28
|
Basov A, Fedulova L, Vasilevskaya E, Trofimova E, Murashova N, Dzhimak S. Sus Scrofa immune tissues as a new source of bioactive substances for skin wound healing. Saudi J Biol Sci 2021; 28:1826-1834. [PMID: 33732068 PMCID: PMC7938156 DOI: 10.1016/j.sjbs.2020.12.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 11/30/2022] Open
Abstract
Influence of a new protein-peptide complex on promoting skin wound healing in male BALB/c mice was studied. Protein-peptide complex, extracted from Sus scrofa immune organs, was percutaneously administered using two methods: by lecithin gel-like liquid crystals and by liquid microemulsion. On the fifth day, wound closure in mice with a linear wound model become faster in group (less 2 days comparison to other ones), which was treated with lecithin liquid crystals carrying the protein-peptide complex. This promoting healing can be caused by resorption of bioactive high-molecular compounds the animal skin. In mice with the linear wound model, the tensile strength of the scars were respectively higher both in mice, treated using lecithin liquid crystals with protein-peptide complex, and in mice, treated using microemulsion containing protein-peptide complex, by 215.4% and 161.5% relative to the animals, which did not receive bioactive substances for wound treatment. It was associated with the regeneratory effects of tissue- and species-specific protein-peptide complexes, including α-thymosin Sus scrofa (C3VVV8_PIG, m/z 3802.8) and other factors, which were described as parts of the new extracted complex. This reveals that percutaneous administration of the complex reliably activates local regenerative processes in animals.
Collapse
Affiliation(s)
- Alexandr Basov
- Kuban State Medical University, Mitrophana Sedina Street, Krasnodar 350063, Russian Federation.,Kuban State University, Stavropolskaya Street, 149, Krasnodar 350040, Russian Federation
| | - Liliya Fedulova
- The V.M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Talalikhina Street, 26, Moscow 109316, Russian Federation
| | - Ekaterina Vasilevskaya
- The V.M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Talalikhina Street, 26, Moscow 109316, Russian Federation
| | - Ekaterina Trofimova
- Mendeleev University of Chemical Technology of Russia, Miusskaya Square, 9, Moscow 125047, Russian Federation
| | - Nataliya Murashova
- Mendeleev University of Chemical Technology of Russia, Miusskaya Square, 9, Moscow 125047, Russian Federation
| | - Stepan Dzhimak
- Kuban State University, Stavropolskaya Street, 149, Krasnodar 350040, Russian Federation.,The V.M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Talalikhina Street, 26, Moscow 109316, Russian Federation
| |
Collapse
|
29
|
Bersanelli M, Giannarelli D, Leonetti A, Buti S, Tiseo M, Nouvenne A, Ticinesi A, Meschi T, Procopio G, Danielli R. The right immune-modulation at the right time: thymosin α1 for prevention of severe COVID-19 in cancer patients. Future Oncol 2021; 17:1097-1104. [PMID: 33538178 PMCID: PMC7874885 DOI: 10.2217/fon-2020-0754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We presented the rationale for the use of thymosin α1 as prophylaxis of severe COVID-19 in cancer patients undergoing active treatment, constituting the background for the PROTHYMOS study, a prospective, multicenter, open-label, Phase II randomized study, currently in its start-up phase (Eudract no. 2020-006020-13). We aim to offer new hope for this incurable disease, especially to frail patient population, such as patients with cancer. The hypothesis of an effective prophylactic approach to COVID-19 would have immediate clinical relevance, especially given the lack of curative approaches. Moreover, in the ‘COVID-19 vaccine race era’ both clinical and biological results coming from the PROTHYMOS trials could even support the rationale for future combinatorial approaches, trying to rise vaccine efficacy in frail individuals. We present scientific evidence in favor of using a drug (thymosin-α1) that modulates the immune system functions to try and prevent severe COVID-19 in cancer patients who are currently receiving anticancer treatment. Thymosin-α1 is produced normally by the body in the thymus, which is present in children but not in adults. Given the better outcomes of SARS-CoV-2 infections in children, we thought that thymosin-α1 could help to protect adults from severe infections as well. In this review, we explain some scientific evidence and the background of our clinical trial, PROTHYMOS, which is investigating this preventive treatment. Our aim is to offer a new hope to these at-risk cancer patients, particularly for the elderly who are at most risk of developing severe COVID-19. Given the lack of approaches that can provide cures to COVID-19, any possibility to prevent severe infection should be explored.
Collapse
Affiliation(s)
- Melissa Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy.,Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Diana Giannarelli
- Biostatistical Unit, Regina Elena National Cancer Institute, IRCCS, Via Elio Chianesi 53, Rome, 00144, Italy
| | - Alessandro Leonetti
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy.,Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy.,Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy.,Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Antonio Nouvenne
- Geriatric Rehabilitation Medical Department, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Andrea Ticinesi
- Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy.,Geriatric Rehabilitation Medical Department, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Tiziana Meschi
- Medicine & Surgery Department, University of Parma, Via Gramsci 14, Parma, 43126, Italy.,Geriatric Rehabilitation Medical Department, University Hospital of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Giuseppe Procopio
- Genito-Urinary Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori of Milan, Via Giacomo Venezian, 1, Milano, 20133, Italy
| | - Riccardo Danielli
- Immuno-Oncology Unit, University Hospital of Siena, Viale Mario Bracci 16, Siena, 53100, Italy
| |
Collapse
|
30
|
Matteucci C, Minutolo A, Balestrieri E, Petrone V, Fanelli M, Malagnino V, Ianetta M, Giovinazzo A, Barreca F, Di Cesare S, De Marco P, Miele MT, Toschi N, Mastino A, Sinibaldi Vallebona P, Bernardini S, Rogliani P, Sarmati L, Andreoni M, Grelli S, Garaci E. Thymosin Alpha 1 Mitigates Cytokine Storm in Blood Cells From Coronavirus Disease 2019 Patients. Open Forum Infect Dis 2021; 8:ofaa588. [PMID: 33506065 PMCID: PMC7798699 DOI: 10.1093/ofid/ofaa588] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) is characterized by immune-mediated lung injury and complex alterations of the immune system, such as lymphopenia and cytokine storm, that have been associated with adverse outcomes underlining a fundamental role of host response in severe acute respiratory syndrome coronavirus 2 infection and the pathogenesis of the disease. Thymosin alpha 1 (Tα1) is one of the molecules used in the management of COVID-19, because it is known to restore the homeostasis of the immune system during infections and cancer. METHODS In this study, we captured the interconnected biological processes regulated by Tα1 in CD8+ T cells under inflammatory conditions. RESULTS Genes associated with cytokine signaling and production were upregulated in blood cells from patients with COVID-19, and the ex vivo treatment with Tα1-mitigated cytokine expression, and inhibited lymphocyte activation in a CD8+ T-cell subset specifically. CONCLUSION These data suggest the potential role of Tα1 in modulating the immune response homeostasis and the cytokine storm in vivo.
Collapse
Affiliation(s)
- Claudia Matteucci
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Antonella Minutolo
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Emanuela Balestrieri
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Vita Petrone
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Marialaura Fanelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Vincenzo Malagnino
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, Italy
| | - Marco Ianetta
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, Italy
| | | | - Filippo Barreca
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, Italy
| | - Silvia Di Cesare
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Unit of Immune and Infectious Diseases, Academic Department of Pediatrics, Bambino Gesù Childrens’ Hospital-Scientific Institute for Research and Healthcare (IRCCS), Rome, Italy
| | - Patrizia De Marco
- Respiratory Medicine Unit, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Martino Tony Miele
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Athinoula A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, Massachusetts, USA
| | - Antonio Mastino
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Paola Sinibaldi Vallebona
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Institute of Translational Pharmacology, National Research Council, Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Respiratory Medicine Unit, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - Loredana Sarmati
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, Italy
| | - Massimo Andreoni
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Infectious Diseases Clinic, Policlinic of Tor Vergata, Rome, Italy
| | - Sandro Grelli
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
- Virology Unit, Policlinic of Tor Vergata, Rome, Italy
| | - Enrico Garaci
- University San Raffaele, Rome, Italy
- IRCCS San Raffaele Pisana, Rome, Italy
| |
Collapse
|
31
|
Binder U, Skerra A. PASylated Thymosin α1: A Long-Acting Immunostimulatory Peptide for Applications in Oncology and Virology. Int J Mol Sci 2020; 22:ijms22010124. [PMID: 33374407 PMCID: PMC7795856 DOI: 10.3390/ijms22010124] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/29/2022] Open
Abstract
Thymosin α1 (Tα1) is an immunostimulatory peptide for the treatment of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections and used as an immune enhancer, which also offers prospects in the context of COVID-19 infections and cancer. Manufacturing of this N-terminally acetylated 28-residue peptide is demanding, and its short plasma half-life limits in vivo efficacy and requires frequent dosing. Here, we combined the PASylation technology with enzymatic in situ N-acetylation by RimJ to produce a long-acting version of Tα1 in Escherichia coli at high yield. ESI-MS analysis of the purified fusion protein indicated the expected composition without any signs of proteolysis. SEC analysis revealed a 10-fold expanded hydrodynamic volume resulting from the fusion with a conformationally disordered Pro/Ala/Ser (PAS) polypeptide of 600 residues. This size effect led to a plasma half-life in rats extended by more than a factor 8 compared to the original synthetic peptide due to retarded kidney filtration. Our study provides the basis for therapeutic development of a next generation thymosin α1 with prolonged circulation. Generally, the strategy of producing an N-terminally protected PASylated peptide solves three major problems of peptide drugs: (i) instability in the expression host, (ii) rapid degradation by serum exopeptidases, and (iii) low bioactivity because of fast renal clearance.
Collapse
Affiliation(s)
- Uli Binder
- XL-protein GmbH, Lise-Meitner-Str. 30, 85354 Freising, Germany
- Correspondence: (U.B.); (A.S.)
| | - Arne Skerra
- Lehrstuhl für Biologische Chemie, Technische Universität München, Emil-Erlenmeyer-Forum 5, 85354 Freising, Germany
- Correspondence: (U.B.); (A.S.)
| |
Collapse
|
32
|
Dominari A, III DH, Pandav K, Matos W, Biswas S, Reddy G, Thevuthasan S, Khan MA, Mathew A, Makkar SS, Zaidi M, Fahem MMM, Beas R, Castaneda V, Paul T, Halpern J, Baralt D. Thymosin alpha 1: A comprehensive review of the literature. World J Virol 2020; 9:67-78. [PMID: 33362999 PMCID: PMC7747025 DOI: 10.5501/wjv.v9.i5.67] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/19/2020] [Accepted: 12/02/2020] [Indexed: 02/06/2023] Open
Abstract
Thymosin alpha 1 is a peptide naturally occurring in the thymus that has long been recognized for modifying, enhancing, and restoring immune function. Thymosin alpha 1 has been utilized in the treatment of immunocompromised states and malignancies, as an enhancer of vaccine response, and as a means of curbing morbidity and mortality in sepsis and numerous infections. Studies have postulated that thymosin alpha 1 could help improve the outcome in severely ill corona virus disease 2019 patients by repairing damage caused by overactivation of lymphocytic immunity and how thymosin alpha 1 could prevent the excessive activation of T cells. In this review, we discuss key literature on the background knowledge and current clinical uses of thymosin alpha 1. Considering the known biochemical properties including antibacterial and antiviral properties, time-honored applications, and the new promising findings regarding the use of thymosin, we believe that thymosin alpha 1 deserves further investigation into its antiviral properties and possible repurposing as a treatment against severe acute respiratory syndrome coronavirus-2.
Collapse
Affiliation(s)
- Asimina Dominari
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Donald Hathaway III
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Krunal Pandav
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Wanessa Matos
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Sharmi Biswas
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Gowry Reddy
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Sindhu Thevuthasan
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Muhammad Adnan Khan
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Anoopa Mathew
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Sarabjot Singh Makkar
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Madiha Zaidi
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | | | - Renato Beas
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Valeria Castaneda
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Trissa Paul
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - John Halpern
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| | - Diana Baralt
- Division of Research and Academic Affairs, Larkin Health System, South Miami, FL 33143, United States
| |
Collapse
|
33
|
Costantini C, van de Veerdonk FL, Romani L. Covid-19-Associated Pulmonary Aspergillosis: The Other Side of the Coin. Vaccines (Basel) 2020; 8:vaccines8040713. [PMID: 33271780 PMCID: PMC7711593 DOI: 10.3390/vaccines8040713] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/19/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
The immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a critical factor in the clinical presentation of COVID-19, which may range from asymptomatic to a fatal, multi-organ disease. A dysregulated immune response not only compromises the ability of the host to resolve the viral infection, but may also predispose the individual to secondary bacterial and fungal infections, a risk to which the current therapeutic immunomodulatory approaches significantly contribute. Among the secondary infections that may occur in COVID-19 patients, coronavirus-associated pulmonary aspergillosis (CAPA) is emerging as a potential cause of morbidity and mortality, although many aspects of the disease still remain unresolved. With this opinion, we present the current view of CAPA and discuss how the same mechanisms that underlie the dysregulated immune response in COVID-19 increase susceptibility to Aspergillus infection. Likewise, resorting to endogenous pathways of immunomodulation may not only restore immune homeostasis in COVID-19 patients, but also reduce the risk for aspergillosis. Therefore, CAPA represents the other side of the coin in COVID-19 and our advances in the understanding and treatment of the immune response in COVID-19 should represent the framework for the study of CAPA.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy;
| | - Frank L. van de Veerdonk
- Department of Internal Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, 06132 Perugia, Italy;
- Correspondence: ; Tel.: +39-075-5858234
| |
Collapse
|
34
|
Bellet MM, Borghi M, Pariano M, Renga G, Stincardini C, D'Onofrio F, Brancorsini S, Garaci E, Costantini C, Romani L. Thymosin alpha 1 exerts beneficial extrapulmonary effects in cystic fibrosis. Eur J Med Chem 2020; 209:112921. [PMID: 33071052 DOI: 10.1016/j.ejmech.2020.112921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/12/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
Cystic fibrosis (CF) is an autosomal recessive disorder caused by mutations in the gene encoding for the ion channel Cystic Fibrosis Transmembrane conductance Regulator (CFTR). Long considered a lung disease for the devastating impact on the respiratory function, the recent diagnostic and therapeutic advances have shed the light on the extra-pulmonary manifestations of CF, including gastrointestinal, hepatobiliary and pancreatic symptoms. We have previously demonstrated that thymosin alpha1 (Tα1), a naturally occurring immunomodulatory peptide, displays multi-sided beneficial effects in CF that concur in ameliorating the lung inflammatory pathology. In the present study, by resorting to murine models of gut inflammation with clinical relevance for CF patients, we demonstrate that Tα1 can also have beneficial effects in extrapulmonary pathology. Specifically, Tα1 restored barrier integrity and immune homeostasis in the inflamed gut of CF mice as well as in mice with the metabolic syndrome, a disorder that may arise in CF patients with high caloric intake despite pancreatic sufficiency. The protective effects of Tα1 also extended to pancreas and liver, further emphasizing the beneficial effects of Tα1 in extra-pulmonary complications of CF. By performing wide-ranging multi-organ anti-inflammatory effects, Tα1 could potentially integrate current therapeutic approaches to tackle the complex symptomatology of CF disease.
Collapse
Affiliation(s)
- Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| | - Monica Borghi
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Claudia Stincardini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Stefano Brancorsini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, 00166, Rome, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, 06132, Italy.
| |
Collapse
|
35
|
Yang Z, Guo J, Cui K, Du Y, Zhao H, Zhu L, Weng L, Tang W, Guo J, Zhang T, Shi X, Zong H, Jin S, Ma W. Thymosin alpha-1 blocks the accumulation of myeloid suppressor cells in NSCLC by inhibiting VEGF production. Biomed Pharmacother 2020; 131:110740. [PMID: 32942159 DOI: 10.1016/j.biopha.2020.110740] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Thymosin alpha-1 (TA) has been reported to inhibit tumor growth as an immunomodulator. However, its mechanism of action in immunosuppressive cells is unclear. The purpose of this study was to investigate whether TA can reshape the immune microenvironment by inhibiting the function of myeloid-derived suppressor cells (MDSCs) in non-small cell lung carcinoma (NSCLC). METHODS The effects of TA on peripheral blood monocytic MDSCs (M-MDSCs) in patients with NSCLC and on the apoptosis and migration of M-MDSCs were studied. A mouse subcutaneous xenograft tumor model was constructed, and the effect of TA on M-MDSC migration was evaluated. Quantitative real-time PCR, Western blotting, flow cytometry and immunohistochemistry were used to examine the mechanism by which TA affects M-MDSCs. RESULTS TA not only promoted the apoptosis of M-MDSCs by reducing the Bcl-2/BAX ratio but also and more importantly inhibited the migration of MDSCs to the tumor microenvironment by suppressing the production of vascular endothelial growth factor (VEGF) through the downregulation of hypoxia-inducible factor (HIF)-1α in tumor cells. CONCLUSIONS TA may have a novel antitumor effect mediated by decreasing M-MDSC accumulation in the tumor microenvironment through reduced VEGF production.
Collapse
Affiliation(s)
- Zhenzhen Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiacheng Guo
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China; Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Kang Cui
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yabing Du
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lili Zhu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Lanling Weng
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Wenxue Tang
- Center for Precision Medicine of Zhengzhou University, Zhengzhou, Henan, 450052, China; Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Jiancheng Guo
- Center for Precision Medicine of Zhengzhou University, Zhengzhou, Henan, 450052, China; Departments of Otolaryngology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450000, China
| | - Tengfei Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaojing Shi
- Laboratory Animal Center, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Hong Zong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Shuiling Jin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Wang Ma
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| |
Collapse
|
36
|
Renga G, Bellet MM, Pariano M, Gargaro M, Stincardini C, D'Onofrio F, Mosci P, Brancorsini S, Bartoli A, Goldstein AL, Garaci E, Romani L, Costantini C. Thymosin α1 protects from CTLA-4 intestinal immunopathology. Life Sci Alliance 2020; 3:3/10/e202000662. [PMID: 32817121 PMCID: PMC7441522 DOI: 10.26508/lsa.202000662] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/21/2022] Open
Abstract
This study demonstrates that Tα1 protects mice from anti–CTLA-4–induced colitis and sustains its antitumor activity, thus suggesting that Tα1 may be used in combination protocols. The advent of immune checkpoint inhibitors has represented a major boost in cancer therapy, but safety concerns are increasingly being recognized. Indeed, although beneficial at the tumor site, unlocking a safeguard mechanism of the immune response may trigger autoimmune-like effects at the periphery, thus making the safety of immune checkpoint inhibitors a research priority. Herein, we demonstrate that thymosin α1 (Tα1), an endogenous peptide with immunomodulatory activities, can protect mice from intestinal toxicity in a murine model of immune checkpoint inhibitor–induced colitis. Specifically, Tα1 efficiently prevented immune adverse pathology in the gut by promoting the indoleamine 2,3-dioxygenase (IDO) 1–dependent tolerogenic immune pathway. Notably, Tα1 did not induce IDO1 in the tumor microenvironment, but rather modulated the infiltration of T-cell subsets by inverting the ratio between CD8+ and Treg cells, an effect that may depend on Tα1 ability to regulate the differentiation and chemokine expression profile of DCs. Thus, through distinct mechanisms that are contingent upon the context, Tα1 represents a plausible candidate to improve the safety/efficacy profile of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Fiorella D'Onofrio
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Paolo Mosci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Andrea Bartoli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Enrico Garaci
- University San Raffaele and Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
37
|
Lunin S, Khrenov M, Glushkova O, Parfenyuk S, Novoselova T, Novoselova E. Precursors of thymic peptides as stress sensors. Expert Opin Biol Ther 2020; 20:1461-1475. [PMID: 32700610 DOI: 10.1080/14712598.2020.1800636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION A large volume of data indicates that the known thymic hormones, thymulin, thymopoietin, thymosin-α, thymosin-β, and thymic humoral factor-y2, exhibit different spectra of activities. Although large in volume, available data are rather fragmented, resulting in a lack of understanding of the role played by thymic hormones in immune homeostasis. AREA COVERED Existing data compartmentalizes the effect of thymic peptides into 2 categories: influence on immune cells and interconnection with neuroendocrine systems. The current study draws attention to a third aspect of the thymic peptide effect that has not been clarified yet, wherein ubiquitous and highly abundant intranuclear precursors of so called 'thymic peptides' play a fundamental role in all somatic cells. EXPERT OPINION Our analysis indicated that, under certain stress-related conditions, these precursors are cleaved to form immunologically active peptides that rapidly leave the nucleus and intracellular spaces, to send 'distress signals' to the immune system, thereby acting as stress sensors. We propose that these peptides may form a link between somatic cells and immune as well as neuroendocrine systems. This model may provide a better understanding of the mechanisms underlying immune homeostasis, leading thereby to the development of new therapeutic regimes utilizing the characteristics of thymic peptides.
Collapse
Affiliation(s)
- Sergey Lunin
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| | - Maxim Khrenov
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| | - Olga Glushkova
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| | - Svetlana Parfenyuk
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| | - Tatyana Novoselova
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| | - E Novoselova
- Laboratory of Reception Mechanisms, Institute of Cell Biophysics of the Russian Academy of Sciences, PSCBR RAS , Pushchino, Russia
| |
Collapse
|
38
|
Calf thymus polypeptide improved hematopoiesis via regulating colony-stimulating factors in BALB/c mice with hematopoietic dysfunction. Int J Biol Macromol 2020; 156:204-216. [PMID: 32156537 DOI: 10.1016/j.ijbiomac.2020.03.041] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/31/2022]
Abstract
Calf thymus polypeptide (CTP) is prepared from calf thymus. It has a molecular mass of <10 kilodalton (kDa) and contains 17 types of amino acids. This study investigated the hematopoietic function-improvement effect of CTP in CHRF, K562, and bone marrow mononuclear cells; mice with immunosuppression; and with hematopoietic dysfunction. In mice with immunosuppression, CTP enhanced the cytotoxic activity of natural killer cells and the proliferation of lymphocytes and regulated the levels of immunoglobulins. It also enhanced the proliferation and differentiation of CHRF and K562 cells by upregulating the expression of proliferation- and differentiation-related proteins. In mice with hematopoietic dysfunction, CTP restored white blood cell, neutrophil, and hemoglobin proportions in the peripheral blood and enhanced the levels of B lymphocytes and hematopoietic stem cells and progenitor cells in the bone marrow. CTP effectively regulated the levels of hematopoiesis-related cytokines, such as granulocyte colony-stimulating factor (G-CSF), macrophage colony-stimulating factor (M-CSF), interleukin 2, and interferons-γ, and enhanced the expression of hematopoiesis-related proteins in both primary bone marrow cells and mice with hematopoietic dysfunction. These results indicate that CTP has hematopoietic function-improvement effect and this effect may be related to the modulation of colony-stimulating factors (CSFs) and related signaling pathways.
Collapse
|
39
|
Aslam MS, Gull I, Mahmood MS, Iqbal MM, Abbas Z, Tipu I, Ahmed A, Athar MA. High yield expression, characterization, and biological activity of IFNα2-Tα1 fusion protein. Prep Biochem Biotechnol 2019; 50:281-291. [PMID: 31718419 DOI: 10.1080/10826068.2019.1689509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The use of interferon α-2 in combination with thymosin α-1 shows higher anti-cancer effect in comparison when both are used individually because of their synergistic effects. In this study we produced an important human interferon α-2-thymosin α-1 (IFNα2-Tα1) fusion protein with probable pharmaceutical properties coupled to its high-level expression, characterization, and study of its biological activity. The IFNα2-Tα1 fusion gene was constructed by over-lap extension PCR and expressed in Escherichia coli expression system. The expression of IFNα2-Tα1 fusion protein was optimized to higher level and its maximum expression was obtained in modified terrific broth medium when lactose was used as inducer. The fusion protein was refolded into its native biologically active form with maximum yield of 83.14% followed by purification with ∼98% purity and 69% final yield. A band of purified IFNα2-Tα1 fusion protein equal to ∼23 kDa was observed on 12 % SDS-PAGE gel. The integrity of IFNα2-Tα1 fusion protein was confirmed by western blot analysis and secondary structure was assessed by CD spectroscopy. When IFNα2-Tα1 fusion protein was subjected to its biological activity analysis it was observed that it exhibits both IFNα2 & Tα1 activities as well as significantly higher anticancer activity as compared to IFNα-2 alone.
Collapse
Affiliation(s)
| | - Iram Gull
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | | | | | - Zaigham Abbas
- Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan
| | - Imran Tipu
- Department of Life Sciences, School of Sciences, University of Management and Technology, Lahore, Pakistan
| | - Aftab Ahmed
- School of Biological Sciences, University of the Punjab, Lahore, Pakistan
| | - Muhammad Amin Athar
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| |
Collapse
|
40
|
Zhang L, Wei X, Zhang R, Petitte JN, Si D, Li Z, Cheng J, Du M. Design and Development of a Novel Peptide for Treating Intestinal Inflammation. Front Immunol 2019; 10:1841. [PMID: 31447849 PMCID: PMC6691347 DOI: 10.3389/fimmu.2019.01841] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/22/2019] [Indexed: 12/24/2022] Open
Abstract
Intestinal inflammatory disorders, such as inflammatory bowel disease (IBD), are associated with increased pro-inflammatory cytokine secretion in the intestines. Furthermore, intestinal inflammation increases the risk of enteric cancer, which is a common malignancy globally. Native anti-inflammatory peptides are a class of anti-inflammatory agents that could be used in the treatment of several intestinal inflammation conditions. However, potential cytotoxicity, and poor anti-inflammatory activity have prevented their development as anti-inflammatory agents. Therefore, in this study, we designed and developed a novel hybrid peptide for the treatment of intestinal inflammation. Eight hybrid peptides were designed by combining the active centers of antimicrobial peptides, including LL-37 (13-36), YW12D, innate defense regulator 1, and cathelicidin 2 (1-13) with thymopentin or the active center of thymosin alpha 1 (Tα1) (17-24). The hybrid peptide, LL-37-Tα1 (LTA), had improved anti-inflammatory activity with minimal cytotoxicity. LTA was screened by molecule docking and in vitro experiments. Likewise, its anti-inflammatory effects and mechanisms were also evaluated using a lipopolysaccharide (LPS)-induced intestinal inflammation murine model. The results showed that LTA prevented LPS-induced impairment in the jejunum epithelium tissues and infiltration of leukocytes, which are both histological markers of inflammation. Additionally, LTA decreased the levels of tumor necrosis factor-alpha, interferon-gamma, interleukin-6, and interleukin-1β. LTA increased the expression of zonula occludens-1 and occludin, and reduced permeability and apoptosis in the jejunum of LPS-treated mice. Additionally, its anti-inflammatory effect is associated with neutralizing LPS, binding to the Toll-like receptor 4-myeloid differentiation factor 2 (TLR4/MD-2) complex, and modulating the nuclear factor-kappa B signal transduction pathway. The findings of this study suggest that LTA may be an effective therapeutic agent in the treatment of intestinal inflammation.
Collapse
Affiliation(s)
- Lulu Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xubiao Wei
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Rijun Zhang
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jim N Petitte
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC, United States
| | - Dayong Si
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhongxuan Li
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Junhao Cheng
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Mengsi Du
- Laboratory of Feed Biotechnology, State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
41
|
Silva LBR, Taira CL, Dias LS, Souza ACO, Nosanchuk JD, Travassos LR, Taborda CP. Experimental Therapy of Paracoccidioidomycosis Using P10-Primed Monocyte-Derived Dendritic Cells Isolated From Infected Mice. Front Microbiol 2019; 10:1727. [PMID: 31417520 PMCID: PMC6685297 DOI: 10.3389/fmicb.2019.01727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/12/2019] [Indexed: 11/13/2022] Open
Abstract
Paracoccidioidomycosis (PCM) is an endemic mycosis in Latin American caused by the thermodimorphic fungi of the genus Paracoccidioides spp. Notably, a Th1 immune response is required to control PCM. In this context, dendritic cells (DCs) seem to be essential players in capture, processing and presentation of Paracoccidioides antigens to naïve T cells and their further activation. We have previously demonstrated that differentiated DCs from bone marrow cells, pulsed with the immunoprotective peptide 10 (P10), effectively control experimental PCM immunocompetent and immunosuppressed mice. However, this procedure may not be infeasible or it is limited for the therapy of human patients. Therefore, we have sought a less invasive but equally effective approach that would better mimics the autologous transplant of DC in a human patient. Here, we isolated and generated monocyte differentiated dendritic cells (MoDCs) from infected mice, pulsed them with P-10, and used them in the therapy of PCM in syngeneic mice. Similar to the results using BMDCs, the P10-pulsed MoDCs stimulated the proliferation of CD4+ T lymphocytes, induced a mixed production of Th1/Th2 cytokines and decreased the fungal burden in murine lungs in the setting of PCM. The process of differentiating MoDCs derived from an infected host, and subsequently used for therapy of PCM is much simpler than that for obtaining BMDCs, and represents a more reasonable approach to treat patients infected with Paracoccidioides. The results presented suggest that P10-primed MoDC may be a promising strategy to combat complicated PCM as well as to significantly shorten the lengthy requirements for treatment of patients with this fungal disease.
Collapse
Affiliation(s)
- Leandro B R Silva
- USP-LIM53, Laboratory of Medical Mycology, Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil
| | - Cleison L Taira
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Lucas S Dias
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ana C O Souza
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Joshua D Nosanchuk
- Departments of Medicine (Division of Infectious Diseases) and Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, United States
| | - Luiz R Travassos
- Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo, São Paulo, Brazil
| | - Carlos P Taborda
- USP-LIM53, Laboratory of Medical Mycology, Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil.,Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
42
|
Ahmad B, Hanif Q, Xubiao W, Lulu Z, Shahid M, Dayong S, Rijun Z. Expression and Purification of Hybrid LL-37Tα1 Peptide in Pichia pastoris and Evaluation of Its Immunomodulatory and Anti-inflammatory Activities by LPS Neutralization. Front Immunol 2019; 10:1365. [PMID: 31258535 PMCID: PMC6587124 DOI: 10.3389/fimmu.2019.01365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022] Open
Abstract
This study pertains to the new approach for the development of hybrid peptide LL-37Tα1 and its biomedical applications. A linear cationic hybrid peptide, LL-37Tα1 was derived from two parental peptides (LL-37 and Tα1) recognized as potent anti-endotoxin without any hemolytic or cytotoxic activity. We successfully cloned the gene of hybrid peptide LL-37Tα1 in PpICZαA vector and expressed in the Pichia pastoris. The recombinant peptide was purified by Ni-affinity column and reverse-phase high performance liquid chromatography (RP-HPLC) with an estimated molecular mass of 3.9 kDa as determined by SDS-PAGE and mass spectrometry. We analyzed the LPS neutralization by limulus amebocyte lysate (LAL) activity and the results indicate that the hybrid peptide LL-37Tα1 directly binds endotoxin and significantly (p < 0.05) neutralizes the effect of LPS in a dose-dependent manner. Lactate dehydrogenase (LDH) assay revealed that LL-37Tα1 successfully reduces the LPS-induced cytotoxicity in mouse RAW264.7 macrophages. Moreover, it significantly (p < 0.05) decreased the levels of nitric oxide, proinflammatory cytokines including TNF-α, IL-6, IL-1β, and diminished the number of apoptotic cells in LPS-stimulated mouse RAW264.7 macrophages. Our results suggest that the P. pastoris expression system is cost-effective for commercial production of the immunomodulatory and anti-inflammatory hybrid peptide (IAHP) LL-37Tα1 and the peptide may serve as effective anti-endotoxin/anti-inflammatory agent with minimal cytotoxicity.
Collapse
Affiliation(s)
- Baseer Ahmad
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Quratulain Hanif
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China.,National Institute for Biotechnology and Genetic Engineering, Pakistan Institute of Engineering and Applied Sciences, Faisalabad, Pakistan
| | - Wei Xubiao
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhang Lulu
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Muhammad Shahid
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China.,College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Si Dayong
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhang Rijun
- State Key Laboratory of Animal Nutrition and Feed Sciences, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
43
|
Mirkov I, Popov Aleksandrov A, Lazovic B, Glamoclija J, Kataranovski M. Usefulness of animal models of aspergillosis in studying immunity against Aspergillus infections. J Mycol Med 2019; 29:84-96. [DOI: 10.1016/j.mycmed.2019.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/28/2018] [Accepted: 01/14/2019] [Indexed: 01/08/2023]
|
44
|
Abstract
INTRODUCTION Sepsis is a life-threatening organ dysfunction caused by host immune response to infection. In this regard, modifying host immune response may help to eliminate systemic infection and restore organ function. Thymosin alpha 1 (Tα1), acting as an immune modulator, exerts great biological influence in activating and restoring the dysregulated immune response for patients with sepsis. AREAS COVERED In this review, we summarize the clinical studies of Tα1 treatment alone and in combination with anti-inflammatory for patients with sepsis or septic shock. Clinical studies were collected from available English and Chinese databases. EXPERT OPINION In previous studies, single or combined treatment with Tα1 reduced the mortality rate of sepsis, improved the expression of HLA-DR on monocyte, and diminished the incidence of secondary infection. Based on the existing studies, Tα1 seems to be a promising alternative adjuvant therapy for sepsis. However, sepsis is a remarkably heterogeneous clinical syndrome, so much so that it is impossible to generalize the clinical results to all septic patients. Also, the present studies on Tα1 treatment are not able to focus on the immunosuppressive individuals. We believe that selecting septic patients with immunosuppression will be more likely to reveal the efficacy of Tα1 in future trials.
Collapse
Affiliation(s)
- Fei Pei
- a Department of Critical Care Medicine , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , Guangdong , China
| | - Xiangdong Guan
- a Department of Critical Care Medicine , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , Guangdong , China
| | - Jianfeng Wu
- a Department of Critical Care Medicine , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , Guangdong , China
| |
Collapse
|
45
|
Pica F, Gaziano R, Casalinuovo IA, Moroni G, Buè C, Limongi D, D'Agostini C, Tomino C, Perricone R, Palamara AT, Sinibaldi Vallebona P, Garaci E. Serum thymosin alpha 1 levels in normal and pathological conditions. Expert Opin Biol Ther 2019; 18:13-21. [PMID: 30063864 DOI: 10.1080/14712598.2018.1474197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Thymosin alpha 1 (Ta1) is a natural occurring peptide hormone that is crucial for the maintenance of the organism homeostasis. It has been chemically synthesized and used in diseases where the immune system is hindered or malfunctioning. AREAS COVERED Many clinical trials investigate the Ta1 effects in patients with cancer, infectious diseases and as a vaccine enhancer. The number of diseases that could benefit from Ta1 treatment is increasing. To date, questions remain about the physiological basal levels of Ta1 and the most effective dose and schedule of treatment. Evidence is growing that diseases characterized by deregulation of immune and/or inflammatory responses are associated with serum levels of Ta1 significantly lower than those of healthy individuals: to date, B hepatitis, psoriatic arthritis, multiple sclerosis and sepsis. The sputum of cystic fibrosis patients contains lower levels of Ta1 than healthy controls. These data are consistent with the role of Ta1 as a regulator of immunity, tolerance and inflammation. EXPERT OPINION Low serum Ta1 levels are predictive and/or associated with different pathological conditions. In case of Ta1 treatment, it is crucial to know the patient's baseline serum Ta1 level to establish effective treatment protocols and monitor their effectiveness over time.
Collapse
Affiliation(s)
- Francesca Pica
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy
| | - Roberta Gaziano
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy
| | | | - Gabriella Moroni
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy
| | - Cristina Buè
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy
| | - Dolores Limongi
- b IRCCS San Raffaele Pisana , Telematic University , Rome , Italy
| | - Cartesio D'Agostini
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy
| | - Carlo Tomino
- b IRCCS San Raffaele Pisana , Telematic University , Rome , Italy
| | - Roberto Perricone
- c Medicine of Systems, Rheumatology, Allergology and Clinical Immunology , University Tor Vergata , Rome , Italy
| | - Anna Teresa Palamara
- b IRCCS San Raffaele Pisana , Telematic University , Rome , Italy.,d Department of Public Health and Infectious Diseases , Sapienza University of Rome , Rome , Italy
| | | | - Enrico Garaci
- a Departments of Experimental Medicine and Surgery , University Tor Vergata , Rome , Italy.,b IRCCS San Raffaele Pisana , Telematic University , Rome , Italy
| |
Collapse
|
46
|
Severa M, Zhang J, Giacomini E, Rizzo F, Etna MP, Cruciani M, Garaci E, Chopp M, Coccia EM. Thymosins in multiple sclerosis and its experimental models: moving from basic to clinical application. Mult Scler Relat Disord 2019; 27:52-60. [PMID: 30317071 PMCID: PMC7104151 DOI: 10.1016/j.msard.2018.09.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/20/2018] [Accepted: 09/30/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) afflicts more than 2.5 million individuals worldwide and this number is increasing over time. Within the past years, a great number of disease-modifying treatments have emerged; however, efficacious treatments and a cure for MS await discovery. Thymosins, soluble hormone-like peptides produced by the thymus gland, can mediate immune and non-immune physiological processes and have gained interest in recent years as therapeutics in inflammatory and autoimmune diseases. METHODS Pubmed was searched with no time constraints for articles using a combination of the keywords "thymosin/s" or "thymus factor/s" AND "multiple sclerosis", mesh terms with no language restriction. RESULTS Here, we review the state-of-the-art on the effects of thymosins on MS and its experimental models. In particular, we describe what is known in this field on the roles of thymosin-α1 (Tα1) and -β4 (Tβ4) as potential anti-inflammatory as well as neuroprotective and remyelinating molecules and their mechanisms of action. CONCLUSION Based on the data that Tα1 and Tβ4 act as anti-inflammatory molecules and as inducers of myelin repair and neuronal protection, respectively, a possible therapeutic application in MS for Tα1 and Tβ4 alone or combined with other approved drugs may be envisaged. This approach is reasonable in light of the current clinical usage of Tα1 and data demonstrating the safety, tolerability and efficacy of Tβ4 in clinical practice.
Collapse
Affiliation(s)
- Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Jing Zhang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Elena Giacomini
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Melania Cruciani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA,Department of Physics, Oakland University, Rochester, MI, USA
| | | |
Collapse
|
47
|
Wang F, Yu T, Zheng H, Lao X. Thymosin Alpha1-Fc Modulates the Immune System and Down-regulates the Progression of Melanoma and Breast Cancer with a Prolonged Half-life. Sci Rep 2018; 8:12351. [PMID: 30120362 PMCID: PMC6097990 DOI: 10.1038/s41598-018-30956-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 08/08/2018] [Indexed: 02/06/2023] Open
Abstract
Thymosin alpha 1 (Tα1) is a biological response modifier that has been introduced into markets for treating several diseases. Given the short serum half-life of Tα1 and the rapid development of Fc fusion proteins, we used genetic engineering method to construct the recombinant plasmid to express Tα1-Fc (Fc domain of human IgG4) fusion protein. A single-factor experiment was performed with different inducers of varying concentrations for different times to get the optimal condition of induced expression. Pure proteins higher than 90.3% were obtained by using 5 mM lactose for 4 h with a final production about 160.4 mg/L. The in vivo serum half-life of Tα1-Fc is 25 h, almost 13 times longer than Tα1 in mice models. Also, the long-acting protein has a stronger activity in repairing immune injury through increasing number of lymphocytes. Tα1-Fc displayed a more effective antitumor activity in the 4T1 and B16F10 tumor xenograft models by upregulating CD86 expression, secreting IFN-γ and IL-2, and increasing the number of tumor-infiltrating CD4+ T and CD8+ T cells. Our study on the novel modified Tα1 with the Fc segment provides valuable information for the development of new immunotherapy in cancer.
Collapse
Affiliation(s)
- Fanwen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, P.R. China
| | - Tingting Yu
- Dongyangguang pharmaceutical r&d co. LTD, Dongguan, 523000, P.R. China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, P.R. China.
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, P.R. China.
| |
Collapse
|
48
|
Wang F, Xu C, Peng R, Li B, Shen X, Zheng H, Lao X. Effect of a C-end rule modification on antitumor activity of thymosin α1. Biochimie 2018; 154:99-106. [PMID: 30096371 DOI: 10.1016/j.biochi.2018.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 08/04/2018] [Indexed: 12/27/2022]
Abstract
Thymosin α1 (Tα1), a hormone containing 28 amino acids, has been approved in several cancer therapies, but the lack of tumor-targeting hinders its full use in tumor treatment. We designed a new peptide by connecting Tα1 and RGDR, generating a product, Tα1-RGDR, where RGDR is located in the C-end with both tumor-homing and cell internalizing properties (C-end rule peptides, a consensus R/KXXR/K motif). This work aimed to study the antitumor and immunological activities of Tα1-RGDR, and its differences compared with the wild-type Tα1. The antitumor and immunological activities of Tα1-RGDR were measured using the B16F10 tumor and immunologic suppression models. Tα1-RGDR treatment led to significant inhibition of tumor growth at a dose at which Tα1 showed a slight effect in the B16F10 tumor growth model. In the immunologic suppression model, Tα1-RGDR shared almost equivalent immunomodulatory effect with Tα1. These results demonstrated the better therapeutic effects after treatment with Tα1-RGDR compared with Tα1. Moreover, both Tα1-RGDR and Tα1 shared a helical conformation in the presence of trifluoroethanol based on CD spectroscopy. Our dock information of Tα1-RGDR when combined with integrin αvβ3 or neuropilin-1 further confirmed previous experimental results. All these findings suggest that Tα1-RGDR might be a useful therapy for tumors by overcoming its wild type limitation of tumor homing.
Collapse
Affiliation(s)
- Fanwen Wang
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Caoying Xu
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Renhao Peng
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Bin Li
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Xutong Shen
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang 24, Nanjing, 210009, China.
| |
Collapse
|
49
|
Garaci E. From thymus to cystic fibrosis: the amazing life of thymosin alpha 1. Expert Opin Biol Ther 2018; 18:9-11. [PMID: 30063868 DOI: 10.1080/14712598.2018.1484447] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Enrico Garaci
- a University San Raffaele and IRCCS San Raffaele , Rome , Italy
| |
Collapse
|
50
|
Mandaliti W, Nepravishta R, Pica F, Vallebona PS, Garaci E, Paci M. Potential mechanism of thymosin-α1-membrane interactions leading to pleiotropy: experimental evidence and hypotheses. Expert Opin Biol Ther 2018; 18:33-42. [DOI: 10.1080/14712598.2018.1456527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Walter Mandaliti
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
| | - Ridvan Nepravishta
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
- School of Pharmacy, East Anglia University, Norwich, UK
| | - Francesca Pica
- Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata”, Rome, Italy
| | | | - Enrico Garaci
- San Raffaele Pisana Scientific Institute for Research, Hospitalization and Health Care, Rome, Italy
| | - Maurizio Paci
- Department of Chemical Sciences and Technologies, University of Rome “Tor Vergata”, Rome, Italy
| |
Collapse
|