1
|
Vitek M, Zvonar Pobirk A, Roškar R, Matjaž MG. Exploiting the potential of in situ forming liquid crystals: development and in vitro performance of long-acting depots for peptide drug thymosin alpha 1 subcutaneous administration. Drug Deliv 2025; 32:2460708. [PMID: 40066714 PMCID: PMC11899226 DOI: 10.1080/10717544.2025.2460708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/06/2024] [Accepted: 01/24/2025] [Indexed: 03/14/2025] Open
Abstract
The fast-growing filed of long-acting depots for subcutaneous (SC) administration holds significant potential to enhance patient adherence to treatment regimens, particularly in the context of chronic diseases. Among them, injectable in situ forming lyotropic liquid crystals (LCCs) consisting of hexagonal mesophases represent an attractive platform due to their remarkable highly ordered microstructure enabling the sustained drug release. These systems are especially relevant for peptide drugs, as their use is limited by their short plasma half-life and inherent poor stability. In this study, we thus aimed to exploit the potential of a liquid crystalline platform for the sustained release of peptide drug thymosin alpha 1 (Tα1), characterized by a short plasma half-life and with that associated twice-weekly SC administration regimen. We initially selected specified ingredients, with ethanol serving to reduce viscosity and stabilize the peptide drug Tα1, lecithin contributing to LCCs formation and stabilization, and glycerol monooleate or glycerol monolinoleate representing the hexagonal LCCs forming matrix material. The selected studied nonaqueous precursor formulations were characterized by suitable rheological properties for SC injection. A convenient and rapid in situ phase transition of precursor formulations to hexagonal LCCs, triggered by water absorption, was successfully accomplished in vitro. Notably, in situ formed LCCs demonstrated sustained release kinetics of the peptide drug Tα1 for up to 2 weeks of in vitro release testing, offering minimized dosing frequency and thus promoting patient adherence. In summary, the newly developed in situ forming liquid crystalline systems represent prospective injectable long-acting depots for SC administration of the peptide drug Tα1.
Collapse
Affiliation(s)
- Mercedes Vitek
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Alenka Zvonar Pobirk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Roškar
- Department of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Mirjam Gosenca Matjaž
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
2
|
Hu K, Liang L, Song J. Development of a ROS-responsive, glutathione-functionalized injectable hydrogel system for controlled drug release. J Biomater Appl 2025:8853282251334208. [PMID: 40209202 DOI: 10.1177/08853282251334208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Oxidative stress arises from an imbalance between excessive production of reactive oxygen species (ROS) and the body's antioxidant defenses. In neurodegenerative diseases, this imbalance leads to ROS accumulation, causing neuronal dysfunction and cell death. Traditional drug therapies often fail to address the dynamic nature of neuroinflammation, limiting their therapeutic efficacy. To overcome this challenge, we have developed an innovative ROS-responsive injectable hydrogel. This hydrogel is designed to detect oxidative stress sensitively and release glutathione in a controlled manner, thereby modulating inflammation and restoring the damaged immune microenvironment to facilitate tissue repair. The hydrogel was synthesized by crosslinking polyvinyl alcohol (PVA) with sodium alginate modified with 3-aminophenylboronic acid (Alg-PBA). We investigated the hydrogel's formation mechanism and analyzed how component variations affect its morphological and rheological properties. Our findings demonstrate that an optimal Alg-PBA to PVA weight ratio of 2:1 yields a hydrogel with superior mechanical strength. Glutathione (GSH) release studies confirmed the hydrogel's pronounced ROS-responsive drug release behavior. Furthermore, biocompatibility assessments revealed that the hydrogel loaded with 100 μg/mL GSH exhibited excellent compatibility and significantly inhibited neuronal apoptosis under oxygen-glucose deprivation (OGD) conditions. This work presents a promising strategy for treating inflammation-related diseases and provides valuable insights for designing next-generation hydrogels that adapt to injury-responsive microenvironments.
Collapse
Affiliation(s)
- Kai Hu
- The First School of Clinical Medicine, Southern Medical University, Wuhan, China
| | - Linlin Liang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jian Song
- The First School of Clinical Medicine, Southern Medical University, Wuhan, China
| |
Collapse
|
3
|
Salvo N, Charles AM, Mohr AM. The Intersection of Trauma and Immunity: Immune Dysfunction Following Hemorrhage. Biomedicines 2024; 12:2889. [PMID: 39767795 PMCID: PMC11673815 DOI: 10.3390/biomedicines12122889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hemorrhagic shock is caused by rapid loss of a significant blood volume, which leads to insufficient blood flow and oxygen delivery to organs and tissues, resulting in severe physiological derangements, organ failure, and death. Physiologic derangements after hemorrhage are due in a large part to the body's strong inflammatory response, which leads to severe immune dysfunction, and secondary complications such as chronic immunosuppression, increased susceptibility to infection, coagulopathy, multiple organ failure, and unregulated inflammation. Immediate management of hemorrhagic shock includes timely control of the source of bleeding, restoring intravascular volume, preferably with whole blood, and prevention of ischemia and organ failure by optimizing tissue oxygenation. However, currently, there are no clinically effective treatments available that can stabilize the immune response to hemorrhage and reinstate homeostatic conditions. In this review, we will discuss what is known about immunologic dysfunction following hemorrhage and potential therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Alicia M. Mohr
- Department of Surgery, Sepsis and Critical Illness Research Center, College of Medicine, University of Florida, 1600 SW Archer Road Box 100108, Gainesville, FL 32610, USA; (N.S.); (A.M.C.)
| |
Collapse
|
4
|
Li H, Lin S, Wang Y, Shi Y, Fang X, Wang J, Cui H, Bian Y, Qi X. Immunosenescence: A new direction in anti-aging research. Int Immunopharmacol 2024; 141:112900. [PMID: 39137628 DOI: 10.1016/j.intimp.2024.112900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
The immune system is a major regulatory system of the body, that is composed of immune cells, immune organs, and related signaling factors. As an organism ages, observable age-related changes in the function of the immune system accumulate in a process described as 'immune aging. Research has shown that the impact of aging on immunity is detrimental, with various dysregulated responses that affect the function of immune cells at the cellular level. For example, increased aging has been shown to result in the abnormal chemotaxis of neutrophils and decreased phagocytosis of macrophages. Age-related diminished functionality of immune cell types has direct effects on host fitness, leading to poorer responses to vaccination, more inflammation and tissue damage, as well as autoimmune disorders and the inability to control infections. Similarly, age impacts the function of the immune system at the organ level, resulting in decreased hematopoietic function in the bone marrow, a gradual deficiency of catalase in the thymus, and thymic atrophy, resulting in reduced production of related immune cells such as B cells and T cells, further increasing the risk of autoimmune disorders in the elderly. As the immune function of the body weakens, aging cells and inflammatory factors cannot be cleared, resulting in a cycle of increased inflammation that accumulates over time. Cumulatively, the consequences of immune aging increase the likelihood of developing age-related diseases, such as Alzheimer's disease, atherosclerosis, and osteoporosis, among others. Therefore, targeting the age-related changes that occur within cells of the immune system might be an effective anti-aging strategy. In this article, we summarize the relevant literature on immune aging research, focusing on its impact on aging, in hopes of providing new directions for anti-aging research.
Collapse
Affiliation(s)
- Hanzhou Li
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China
| | - Shan Lin
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuming Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuexuan Shi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xixing Fang
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jida Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huantian Cui
- Yunnan University of Chinese Medicine, Yunnan, China.
| | - Yuhong Bian
- Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Xin Qi
- Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Union Medical Center, Tianjin, China.
| |
Collapse
|
5
|
Öhlander A, Lüdtke C, Sahakjan A, Johnsson RE. N-Butylpyrrolidinone is an equally good solvent as N,N-dimethylformamide for microwave assisted solid phase peptide synthesis. J Pept Sci 2024; 30:e3612. [PMID: 38720008 DOI: 10.1002/psc.3612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 10/12/2024]
Abstract
Solid-phase peptide synthesis (SPPS) is the prevailing method for synthesizing research peptides today. However, SPPS is associated with a significant environmental concern due to the utilization of hazardous solvents such as N,N-dimethylformamide (DMF) or N-methylpyrrolidone, which generate substantial waste. In light of this, our research endeavors to identify more environmentally friendly solvents for SPPS. In this study, we have assessed the suitability of five green solvents as alternatives to DMF in microwave assisted SPPS. The solvents evaluated include Cyrene, ethyl acetate, 1,3-dioxolane, tetrahydro-2-methylfuran, and N-Butylpyrrolidinone (NBP). Our investigation encompassed all stages of the synthesis process, from resin swelling, dissolution of reagents, culminating in the successful synthesis of five diverse peptides, including the challenging ACP 65-74, Peptide 18A, Thymosin α1, and Jung-Redemann peptide. Our findings indicate that NBP emerged as a strong contender, performing on par with DMF in all tested syntheses. Furthermore, we observed that combinations of NBP with either ethyl acetate or tetrahydro-2-methylfuran demonstrated excellent results. This research contributes to the pursuit of more sustainable and environmentally conscious practices in peptide synthesis.
Collapse
|
6
|
Cui C, Huo Q, Xiong X, Na S, Mitsuda M, Minami K, Li B, Yokota H. P18: Novel Anticancer Peptide from Induced Tumor-Suppressing Cells Targeting Breast Cancer and Bone Metastasis. Cancers (Basel) 2024; 16:2230. [PMID: 38927935 PMCID: PMC11202002 DOI: 10.3390/cancers16122230] [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: 04/30/2024] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND The skeletal system is a common site for metastasis from breast cancer. In our prior work, we developed induced tumor-suppressing cells (iTSCs) capable of secreting a set of tumor-suppressing proteins. In this study, we examined the possibility of identifying anticancer peptides (ACPs) from trypsin-digested protein fragments derived from iTSC proteomes. METHODS The efficacy of ACPs was examined using an MTT-based cell viability assay, a Scratch-based motility assay, an EdU-based proliferation assay, and a transwell invasion assay. To evaluate the mechanism of inhibitory action, a fluorescence resonance energy transfer (FRET)-based GTPase activity assay and a molecular docking analysis were conducted. The efficacy of ACPs was also tested using an ex vivo cancer tissue assay and a bone microenvironment assay. RESULTS Among the 12 ACP candidates, P18 (TDYMVGSYGPR) demonstrated the most effective anticancer activity. P18 was derived from Arhgdia, a Rho GDP dissociation inhibitor alpha, and exhibited inhibitory effects on the viability, migration, and invasion of breast cancer cells. It also hindered the GTPase activity of RhoA and Cdc42 and downregulated the expression of oncoproteins such as Snail and Src. The inhibitory impact of P18 was additive when it was combined with chemotherapeutic drugs such as Cisplatin and Taxol in both breast cancer cells and patient-derived tissues. P18 had no inhibitory effect on mesenchymal stem cells but suppressed the maturation of RANKL-stimulated osteoclasts and mitigated the bone loss associated with breast cancer. Furthermore, the P18 analog modified by N-terminal acetylation and C-terminal amidation (Ac-P18-NH2) exhibited stronger tumor-suppressor effects. CONCLUSIONS This study introduced a unique methodology for selecting an effective ACP from the iTSC secretome. P18 holds promise for the treatment of breast cancer and the prevention of bone destruction by regulating GTPase signaling.
Collapse
Affiliation(s)
- Changpeng Cui
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (C.C.); (Q.H.); (X.X.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Qingji Huo
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (C.C.); (Q.H.); (X.X.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Xue Xiong
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (C.C.); (Q.H.); (X.X.)
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
| | - Masaru Mitsuda
- Frontier Research Institute, Chubu University, Aichi 487-8501, Japan;
| | - Kazumasa Minami
- Department of Radiation Oncology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan;
| | - Baiyan Li
- Department of Pharmacology, School of Pharmacy, Harbin Medical University, Harbin 150081, China; (C.C.); (Q.H.); (X.X.)
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA;
- Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
7
|
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
|
8
|
Wixler V, Leite Dantas R, Varga G, Boergeling Y, Ludwig S. Small Spleen Peptides (SSPs) Shape Dendritic Cell Differentiation through Modulation of Extracellular ATP Synthesis Profile. Biomolecules 2024; 14:469. [PMID: 38672485 PMCID: PMC11047987 DOI: 10.3390/biom14040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Restoring peripheral immune tolerance is crucial for addressing autoimmune diseases. An ancient mechanism in maintaining the balance between inflammation and tolerance is the ratio of extracellular ATP (exATP) and adenosine. Our previous research demonstrated the effectiveness of small spleen peptides (SSPs) in inhibiting psoriatic arthritis progression, even in the presence of the pro-inflammatory cytokine TNFα, by transforming dendritic cells (DCs) into tolerogenic cells and fostering regulatory Foxp3+ Treg cells. Here, we identified thymosins as the primary constituents of SSPs, but recombinant thymosin peptides were less efficient in inhibiting arthritis than SSPs. Since Tβ4 is an ecto-ATPase-binding protein, we hypothesized that SSPs regulate exATP profiles. Real-time investigation of exATP levels in DCs revealed that tolerogenic stimulation led to robust de novo exATP synthesis followed by significant degradation, while immunogenic stimulation resulted in a less pronounced increase in exATP and less effective degradation. These contrasting exATP profiles were crucial in determining whether DCs entered an inflammatory or tolerogenic state, highlighting the significance of SSPs as natural regulators of peripheral immunological tolerance, with potential therapeutic benefits for autoimmune diseases. Finally, we demonstrated that the tolerogenic phenotype of SSPs is mainly influenced by adenosine receptors, and in vivo administration of SSPs inhibits psoriatic skin inflammation.
Collapse
Affiliation(s)
- Viktor Wixler
- Institute of Molecular Virology, Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, Von-Esmarch-Str. 56, 48149 Muenster, Germany; (R.L.D.); (Y.B.); (S.L.)
| | - Rafael Leite Dantas
- Institute of Molecular Virology, Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, Von-Esmarch-Str. 56, 48149 Muenster, Germany; (R.L.D.); (Y.B.); (S.L.)
| | - Georg Varga
- Department of Pediatric Rheumatology and Immunology, University Children’s Hospital Muenster, 48149 Muenster, Germany;
| | - Yvonne Boergeling
- Institute of Molecular Virology, Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, Von-Esmarch-Str. 56, 48149 Muenster, Germany; (R.L.D.); (Y.B.); (S.L.)
| | - Stephan Ludwig
- Institute of Molecular Virology, Center for Molecular Biology of Inflammation (ZMBE), Westfaelische Wilhelms-University, Von-Esmarch-Str. 56, 48149 Muenster, Germany; (R.L.D.); (Y.B.); (S.L.)
| |
Collapse
|
9
|
Bode C, Weis S, Sauer A, Wendel-Garcia P, David S. Targeting the host response in sepsis: current approaches and future evidence. Crit Care 2023; 27:478. [PMID: 38057824 PMCID: PMC10698949 DOI: 10.1186/s13054-023-04762-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
Sepsis, a dysregulated host response to infection characterized by organ failure, is one of the leading causes of death worldwide. Disbalances of the immune response play an important role in its pathophysiology. Patients may develop simultaneously or concomitantly states of systemic or local hyperinflammation and immunosuppression. Although a variety of effective immunomodulatory treatments are generally available, attempts to inhibit or stimulate the immune system in sepsis have failed so far to improve patients' outcome. The underlying reason is likely multifaceted including failure to identify responders to a specific immune intervention and the complex pathophysiology of organ dysfunction that is not exclusively caused by immunopathology but also includes dysfunction of the coagulation system, parenchymal organs, and the endothelium. Increasing evidence suggests that stratification of the heterogeneous population of septic patients with consideration of their host response might led to treatments that are more effective. The purpose of this review is to provide an overview of current studies aimed at optimizing the many facets of host response and to discuss future perspectives for precision medicine approaches in sepsis.
Collapse
Affiliation(s)
- Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
| | - Sebastian Weis
- Institute for Infectious Disease and Infection Control, University Hospital Jena, Friedrich-Schiller University Jena, Jena, Germany
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Jena, Friedrich-Schiller University Jena, Jena, Germany
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll Institute-HKI, Jena, Germany
| | - Andrea Sauer
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Pedro Wendel-Garcia
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Sascha David
- Institute of Intensive Care Medicine, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
10
|
Soeroto AY, Suryadinata H, Yanto TA, Hariyanto TI. The efficacy of thymosin alpha-1 therapy in moderate to critical COVID-19 patients: a systematic review, meta-analysis, and meta-regression. Inflammopharmacology 2023; 31:3317-3325. [PMID: 37845598 DOI: 10.1007/s10787-023-01354-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Effort to search for the optimal COVID-19 treatment has continuously been attempted. Thymosin alpha-1 have immunomodulatory properties which may be beneficial in case of viral infection. This study's goal is to determine whether thymosin alpha-1 is effective in treating people with moderate-to-severe COVID-19. METHODS We searched for literature in 4 database: Scopus, Europe PMC, Medline, ClinicalTrials.gov, and Cochrane Library until March 25th, 2023. If those articles have data on the efficacy of thymosin alpha-1 therapy on COVID-19, they would be included. Risk ratio (RR) and Mean Difference (MD) along with their 95% confidence intervals were used to pool the results of dichotomous and continuous variables, respectively. RESULTS Pooled data from 8 studies indicated that moderate to critical Covid-19 patients who were receiving thymosin alpha-1 therapy had significantly lower mortality from COVID-19 (RR 0.59; 95% CI 0.37-0.93, p = 0.02, I2 = 84%), but without any difference in the needs for mechanical ventilation (RR 0.83; 95% CI 0.48-1.44, p = 0.51, I2 = 74%) and hospital length of stay (MD 2.32; 95% CI - 0.93, 5.58, p = 0.16, I2 = 94%) compared to placebo. The benefits of thymosin alpha-1 on the mortality rate were significantly affected only by sample size (p = 0.0000) and sex (p = 0.0117). CONCLUSION Our study suggests that treatment with thymosin alpha-1 may reduce mortality rate in moderate to critical COVID-19 patients. Randomized clinical trials (RCTs) are still required to verify the findings of our study.
Collapse
Affiliation(s)
- Arto Yuwono Soeroto
- Division of Pulmonology and Critical Illness, Department of Internal Medicine, Padjadjaran University, Bandung, West Java, 45363, Indonesia
| | - Hendarsyah Suryadinata
- Division of Pulmonology and Critical Illness, Department of Internal Medicine, Padjadjaran University, Bandung, West Java, 45363, Indonesia
| | - Theo Audi Yanto
- Faculty of Medicine, Department of Internal Medicine, Pelita Harapan University, Karawaci, Tangerang, 15811, Indonesia
| | - Timotius Ivan Hariyanto
- Faculty of Medicine, Pelita Harapan University, Boulevard Jendral Sudirman Street, Karawaci, Tangerang, 15811, Indonesia.
| |
Collapse
|
11
|
Quagliata M, Papini AM, Rovero P. Therapeutic applications of thymosin peptides: a patent landscape 2018-present. Expert Opin Ther Pat 2023; 33:865-873. [PMID: 38131310 DOI: 10.1080/13543776.2023.2298833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION Thymosins are small proteins found mainly in the thymus. They are involved in several biological processes, including immunoregulation, angiogenesis, and anti-inflammatory activity. Due to these multiple activities, thymosins are widely used as therapeutics. In fact, these peptides have shown interesting results in the treatment of eye disorders, anticancer therapy, and dysregulated immune disorders. AREA COVERED We analyzed the thymosins therapeutic patent landscape describing the most significant patents published after 2018 and originally written in English, classified according to the different type of functions and diseases. We searched 'Thymosin' on Patentscope and Espacenet. EXPERT OPINION Thymalfasin (Zadaxin) is the only FDA-approved thymosine-based drug used to treat chronic hepatitis B and C and as a chemotherapy inducer. This outcome demonstrates how thymosins can be exploited as therapeutics, especially in immunological and anti-cancer therapies. However, the development of modified thymosins could expand their therapeutic interest and application in different diseases. In fact, by chemical modifications, it is possible to increase proteolytic stability in the biological environment, enhance cell permeability, and stabilize the secondary structure of the peptide. Finally, the development of shorter sequences could reduce the cost and production time of these thymosin-based drugs.
Collapse
Affiliation(s)
- Michael Quagliata
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Anna Maria Papini
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of Chemistry "Ugo Schiff", University of Florence, Sesto Fiorentino, Italy
| | - Paolo Rovero
- Interdepartmental Research Unit of Peptide and Protein Chemistry and Biology, Department of NeuroFarBa, University of Florence, Sesto Fiorentino, Italy
| |
Collapse
|
12
|
Wu Y, He L, Guo Y, Wang N. Risk Factors and Drug Efficacy for Severe Illness in Hemodialysis Patients Infected with the Omicron Variant of COVID-19. Kidney Blood Press Res 2023; 48:642-651. [PMID: 37751729 PMCID: PMC10614566 DOI: 10.1159/000534192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/11/2023] [Indexed: 09/28/2023] Open
Abstract
INTRODUCTION The Omicron variant of the novel coronavirus (COVID-19) has been spreading more rapidly and is more infectious, posing a higher risk of death and treatment difficulty for patients undergoing hemodialysis. This study aims to explore the severity rate and risk factors for hemodialysis patients infected with the Omicron variant and to conduct a preliminary analysis of the clinical efficacy of drugs. METHODS Clinical and biochemical indicators of 219 hemodialysis patients infected with the Omicron variant were statistically analyzed. The patients were divided into two groups based on whether they were severely ill or not, and multiple regression analysis was conducted to determine the risk factors for severe illness. The severely ill patients were then grouped based on discharge or death, and the treatment drugs were included as influencing factors for multiple regression analysis to determine the risk factors and protective factors for death of severely ill patients, and drug efficacy analysis was conducted. RESULTS Analysis showed that diabetes, low oxygen saturation, and high C-reactive protein (CRP) were independent risk factors for severe illness in hemodialysis patients infected with the Omicron variant. A history of diabetes and high C-reactive significantly increased the risk of severe illness in patients (aOR: 1.450; aOR: 1.011), while a high oxygen saturation level can reduce this risk (aOR: 0.871). In addition, respiratory distress was an independent risk factor for death in severely patients, significantly reducing the probability of discharge for patients (aOR: 0.152). The drugs thymalfasin and Tanreqing significantly increased the probability of discharge for patients (aOR: 1.472; aOR: 3.104), with the latter having a higher correlation, but with a relatively longer effective course. CONCLUSION Hemodialysis patients infected with the Omicron variant of COVID-19 should pay special attention to their history of diabetes, CRP, and oxygen saturation levels, as well as respiratory distress symptoms, to reduce the risk of severe illness and death. In addition, thymalfasin and Tanreqing may be considered in treatment.
Collapse
Affiliation(s)
- Yan Wu
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,
| | - Lingling He
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongping Guo
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Niansong Wang
- Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Wang Z, Wang C, Fei X, Wu H, Niu P, Shen C. Thymalfasin therapy accelerates COVID-19 pneumonia rehabilitation through anti-inflammatory mechanisms. Pneumonia (Nathan) 2023; 15:14. [PMID: 37743481 PMCID: PMC10518946 DOI: 10.1186/s41479-023-00116-6] [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: 05/15/2023] [Accepted: 08/03/2023] [Indexed: 09/26/2023] Open
Abstract
INTRODUCTION Thymosin drugs are commonly used for the treatment of viral infections due to their immunomodulatory effects. The comprehensive clinical efficacy of Thymalfasin therapy for COVID-19 associated pneumonia is not yet fully researched, another issue, whether the use of thymosin drugs can reduce the rate of COVID-19 progression to severe pneumonia has not been well documented. The aim of the present study was to multi-angle evaluate the clinical efficacy of Thymalfasin therapy for COVID-19 pneumonia by retrospective review of the clinical data of 338 inpatients with common COVID-19 infection who received treatment in our hospital. METHODS The primary index of observation was whether progression to severe pneumonia occurred within a week after admission, and the secondary indexes were the length of hospital stay, time of negative conversion of COVID-19 antigen, the number of peripheral lymphocytes and white blood cells (WBC), and C-reactive protein (CRP) and procalcitonin (PCT) levels,and the control of pneumonia related symptoms, for example, fever, listlessness, inflammatory exudate area shown on lung CT (%). RESULTS The length of hospital stay of patients in Thymalfasin group was significantly shorter than that of patients in the control group (p < 0.01). The proportion of relief of pneumonia related symptoms (fever, fatigue) in the Thymalfasin therapy group was significantly higher than that in the control group, and the inflammatory exudate area shown on CT was significantly lower than that in the control group (p < 0.05). Multivariate logistic regression analysis showed that the use of Thymalfasin was an independent protective factor affecting the progression to severe pneumonia. Multifactorial Cox model analysis indicated that negative conversion of COVID-19 antigen was significantly faster in patients using Thymalfasin and younger patients. CONCLUSION Thymalfasin therapy has shown excellent clinical efficacy in the treatment of COVID-19 pneumonia, it can reduce inflammatory reactions, promote the relief of COVID-19 pneumonia related symptoms such as fever and fatigue, facilitate effusion absorption, and accelerate COVID-19 pneumonia recovery. Thymalfasin can prevent progression of common COVID-19 infection to severe pneumonia via multiple immunity-enhancing and anti-inflammatory protective mechanisms.
Collapse
Affiliation(s)
- Zirui Wang
- Department of Respiratory and Critical Medicine, the Fifth People's Hospital of Wujiang District, Suzhou, 215211, JS, China
| | - Cong Wang
- Department of Respiratory and Critical Medicine, the Fifth People's Hospital of Wujiang District, Suzhou, 215211, JS, China
| | - Xiaohua Fei
- Information Centre, the Fifth People's Hospital of Wujiang District, Suzhou, 215211, JS, China
| | - Haixing Wu
- Department of Pharmacy, the Fifth People's Hospital of Wujiang District, Suzhou, 215211, JS, China
| | - Peiqin Niu
- Department of Medical Record Statistics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Changxing Shen
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072, Shanghai, China.
| |
Collapse
|
14
|
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
|
15
|
Savino W, Lepletier A. Thymus-derived hormonal and cellular control of cancer. Front Endocrinol (Lausanne) 2023; 14:1168186. [PMID: 37529610 PMCID: PMC10389273 DOI: 10.3389/fendo.2023.1168186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 06/26/2023] [Indexed: 08/03/2023] Open
Abstract
The thymus gland is a central lymphoid organ in which developing T cell precursors, known as thymocytes, undergo differentiation into distinct type of mature T cells, ultimately migrating to the periphery where they exert specialized effector functions and orchestrate the immune responses against tumor cells, pathogens and self-antigens. The mechanisms supporting intrathymic T cell differentiation are pleiotropically regulated by thymic peptide hormones and cytokines produced by stromal cells in the thymic microenvironment and developing thymocytes. Interestingly, in the same way as T cells, thymic hormones (herein exemplified by thymosin, thymulin and thymopoietin), can circulate to impact immune cells and other cellular components in the periphery. Evidence on how thymic function influences tumor cell biology and response of patients with cancer to therapies remains unsatisfactory, although there has been some improvement in the knowledge provided by recent studies. Herein, we summarize research progression in the field of thymus-mediated immunoendocrine control of cancer, providing insights into how manipulation of the thymic microenvironment can influence treatment outcomes, including clinical responses and adverse effects of therapies. We review data obtained from clinical and preclinical cancer research to evidence the complexity of immunoendocrine interactions underpinning anti-tumor immunity.
Collapse
Affiliation(s)
- Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Brazilian National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Rio de Janeiro Research Network on Neuroinflammation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- INOVA-IOC Network on Neuroimmunomodulation, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ailin Lepletier
- Institute for Glycomics, Griffith University, Gold Coast, QLD, Australia
| |
Collapse
|
16
|
Wang S, Wei W, Yong H, Zhang Z, Zhang X, Zhang X, Wang S. Synergistic anti-cancer and attenuation effects of thymosin on chemotherapeutic drug vinorelbine in tumor-bearing zebrafish. Biomed Pharmacother 2023; 162:114633. [PMID: 37018994 DOI: 10.1016/j.biopha.2023.114633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/18/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
Vinorelbine, the standard chemotherapy drug on advanced lung cancer, causes adverse events such as immunosuppression and bone marrow suppression. Thus, it is necessary to find drugs that could improve immune function and synergistically enhance the anti-tumor effect of vinorelbine. Thymosin is reported to inhibit tumor growth as an immunomodulator. Herein, to study the synergistic anti-cancer and attenuation effects of thymosin on vinorelbine, human lung cancer A549 cells that were labeled with CM-DiI were transplanted into zebrafish to establish the lung cancer xenotransplanted model. After treatment of vinorelbine and different concentrations of thymosin, the fluorescence intensity of CM-DiI-labeled A549 cells and the number of apoptotic muscle cells in the tumor-bearing zebrafish were detected. Besides, effects of thymosin on vinorelbine-reduced macrophages and T cells were identified in the transgenic zebrafish (Tg:zlyz-EGFP and Tg:rag2-DsRed). Then, the qRT-PCR was used to determine the alterations of the immune-related factors at the transcription level. Thymosin showed a marked synergistic anti-cancer effect with vinorelbine for the xenograft human lung cancer A549 cells, and the synergistic effect enhanced in a dose-dependent manner. Moreover, thymosin alleviated vinorelbine-induced muscle cell apoptosis, macrophage reduction, and T cell suppression. Compared with the vinorelbine group, co-administration with thymosin raised the mRNA levels of TNF-α, TNF-β, INF-γ, and GM-CSF. Thus, thymosin possesses synergistic anti-cancer effect on vinorelbine, and has protective effect on vinorelbine-induced immunosuppression. Thymosin, as an adjuvant immunomodulatory therapy, has great potential in enhancing the clinical application of vinorelbine.
Collapse
|
17
|
Marques A, Torre C, Pinto R, Sepodes B, Rocha J. Treatment Advances in Sepsis and Septic Shock: Modulating Pro- and Anti-Inflammatory Mechanisms. J Clin Med 2023; 12:2892. [PMID: 37109229 PMCID: PMC10142733 DOI: 10.3390/jcm12082892] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/10/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Sepsis is currently defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection, and it affects over 25 million people every year. Even more severe, septic shock is a subset of sepsis defined by persistent hypotension, and hospital mortality rates are higher than 40%. Although early sepsis mortality has greatly improved in the past few years, sepsis patients who survive the hyperinflammation and subsequent organ damage often die from long-term complications, such as secondary infection, and despite decades of clinical trials targeting this stage of the disease, currently, no sepsis-specific therapies exist. As new pathophysiological mechanisms have been uncovered, immunostimulatory therapy has emerged as a promising path forward. Highly investigated treatment strategies include cytokines and growth factors, immune checkpoint inhibitors, and even cellular therapies. There is much to be learned from related illnesses, and immunotherapy trials in oncology, as well as the recent COVID-19 pandemic, have greatly informed sepsis research. Although the journey ahead is a long one, the stratification of patients according to their immune status and the employment of combination therapies represent a hopeful way forward.
Collapse
Affiliation(s)
- Adriana Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Carla Torre
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - Rui Pinto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
- Joaquim Chaves Saúde, Joaquim Chaves Laboratório de Análises Clínicas, Miraflores, 1495-069 Algés, Portugal
| | - Bruno Sepodes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| | - João Rocha
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal; (A.M.); (C.T.); (R.P.); (B.S.)
| |
Collapse
|
18
|
Ke L, Mao W, Shao F, Zhou J, Xu M, Chen T, Liu Y, Tong Z, Windsor J, Ma P, Li W, Chinese Acute Pancreatitis Clinical Trials Group (CAPCTG) KeLuMaoWenjianZhouJingXuMinyiJiangWendiZhangHeLinJiajiaLuMengjieChenYanMaMingminLiGangYeBoLiBaiqiangTongZhihuiLiuYuxiuLiWeiqinShaoFangChenTaoLvNonghuaZhuYinXiaLiangHeWenhuaZhenpingChenPanXintingZhuQingyunWanYoudongMeiHongLiKangChenMiaoHeChengjianYaoHongyiZhuZiguiGuWeiliLuWeihuaWuJingyiZhouFengTuShuminFuLongXueBinggNiHaibinHuangXiaofeiZhouDandanZhangGuoxiuRenLeningLiDahuanZhaoXiangyangZhaoWeiChenXiaomeiSunJunliXinKekeChenWeiweiXuQingchengSongJingchunZengQingboShaoMinZhaoDongshengTuJianfengYangHonggup. 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
|
19
|
Ricci D, Etna MP, Severa M, Fiore S, Rizzo F, Iannetta M, Andreoni M, Balducci S, Stefanelli P, Palamara AT, Coccia EM. Novel evidence of Thymosin α1 immunomodulatory properties in SARS-CoV-2 infection: Effect on innate inflammatory response in a peripheral blood mononuclear cell-based in vitro model. Int Immunopharmacol 2023; 117:109996. [PMID: 36933449 PMCID: PMC10008813 DOI: 10.1016/j.intimp.2023.109996] [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: 01/09/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023]
Abstract
The peculiar property of Thymosin alpha 1 (Tα1) to act as master regulator of immune homeostasis has been successfully defined in different physiological and pathological contexts ranging from cancer to infection. Interestingly, recent papers also demonstrated its mitigating effect on the "cytokine storm" as well as on the T-cell exhaustion/activation in SARS-CoV-2 infected individuals. Nevertheless, in spite of the increasing knowledge on Tα1-induced effects on T cell response confirming the distinctive features of this multifaceted peptide, little is known on its effects on innate immunity during SARS-CoV-2 infection. Here, we interrogated peripheral blood mononuclear cell (PBMC) cultures stimulated with SARS-CoV-2 to disclose Tα1 properties on the main cell players of early response to infection, namely monocytes and myeloid dendritic cells (mDC). Moving from ex vivo data showing an enhancement in the frequency of inflammatory monocytes and activated mDC in COVID-19 patients, a PBMC-based experimental setting reproduced in vitro a similar profile with an increased percentage of CD16+ inflammatory monocytes and mDC expressing CD86 and HLA-DR activation markers in response to SARS-CoV-2 stimulation. Interestingly, the treatment of SARS-CoV-2-stimulated PBMC with Tα1 dampened the inflammatory/activation status of both monocytes and mDC by reducing the release of pro-inflammatory mediators, including TNF-α, IL-6 and IL-8, while promoting the production of the anti-inflammatory cytokine IL-10. This study further clarifies the working hypothesis on Tα1 mitigating action on COVID-19 inflammatory condition. Moreover, these evidence shed light on inflammatory pathways and cell types involved in acute SARS-CoV-2 infection and likely targetable by newly immune-regulating therapeutic approaches.
Collapse
Affiliation(s)
- Daniela Ricci
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy; Roma Tre University, Rome, Italy
| | - Marilena Paola Etna
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Severa
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Stefano Fiore
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Fabiana Rizzo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Marco Iannetta
- Department of System Medicine, Infectious Disease Unit, University of Rome Tor Vergata, Rome, Italy
| | - Massimo Andreoni
- Department of System Medicine, Infectious Disease Unit, University of Rome Tor Vergata, Rome, Italy
| | | | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | | |
Collapse
|
20
|
Pozo-Balado MDM, Bulnes-Ramos Á, Olivas-Martínez I, Garrido-Rodríguez V, Lozano C, Álvarez-Ríos AI, Sánchez-Sánchez B, Sánchez-Bejarano E, Maldonado-Calzado I, Martín-Lara JM, Santamaría JA, Bernal R, González-Escribano MF, Leal M, Pacheco YM. Higher plasma levels of thymosin-α1 are associated with a lower waning of humoral response after COVID-19 vaccination: an eight months follow-up study in a nursing home. Immun Ageing 2023; 20:9. [PMID: 36879319 PMCID: PMC9986663 DOI: 10.1186/s12979-023-00334-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
BACKGROUND Older people achieve lower levels of antibody titers than younger populations after Covid-19 vaccination and show a marked waning humoral immunity over time, likely due to the senescence of the immune system. Nevertheless, age-related predictive factors of the waning humoral immune response to the vaccine have been scarcely explored. In a cohort of residents and healthcare workers from a nursing home that had received two doses of the BNT162b2 vaccine, we measured specific anti-S antibodies one (T1), four (T4), and eight (T8) months after receiving the second dose. Thymic-related functional markers, including thymic output, relative telomere length, and plasma thymosin-α1 levels, as well as immune cellular subsets, and biochemical and inflammatory biomarkers, were determined at T1, and tested for their associations with the magnitude of the vaccine response (T1) and the durability of such response both, at the short- (T1-T4) and the long-term (T1-T8). We aimed to identify age-related factors potentially associated with the magnitude and persistence of specific anti-S immunoglobulin G (IgG)-antibodies after COVID-19 vaccination in older people. RESULTS Participants (100% men, n = 98), were subdivided into three groups: young (< 50 years-old), middle-age (50-65 years-old), and older (≥65 years-old). Older participants achieved lower antibody titers at T1 and experienced higher decreases in both the short- and long-term. In the entire cohort, while the magnitude of the initial response was mainly associated with the levels of homocysteine [β (95% CI); - 0.155 (- 0.241 to - 0.068); p = 0.001], the durability of such response at both, the short-term and the long-term were predicted by the levels of thymosin-α1 [- 0.168 (- 0.305 to - 0.031); p = 0.017, and - 0.123 (- 0.212 to - 0.034); p = 0.008, respectively]. CONCLUSIONS Higher plasma levels of thymosin-α1 were associated with a lower waning of anti-S IgG antibodies along the time. Our results suggest that plasma levels of thymosin-α1 could be used as a biomarker for predicting the durability of the responses after COVID-19 vaccination, possibly allowing to personalize the administration of vaccine boosters.
Collapse
Affiliation(s)
- María Del Mar Pozo-Balado
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain
| | - Ángel Bulnes-Ramos
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain
| | - Israel Olivas-Martínez
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain
| | - Vanesa Garrido-Rodríguez
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain
| | - Carmen Lozano
- Microbiology Service, Virgen del Rocío University Hospital (HUVR), Seville, Spain
| | - Ana I Álvarez-Ríos
- Biochemistry Service, Virgen del Rocío University Hospital (HUVR), Seville, Spain
| | | | | | | | | | | | - Rafael Bernal
- Hogar Residencia de la Santa Caridad, Seville, Spain
| | - María Francisca González-Escribano
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain.,Immunology Service, Virgen del Rocío University Hospital (HUVR), Seville, Spain
| | - Manuel Leal
- Hogar Residencia de la Santa Caridad, Seville, Spain.,Internal Medicine Service, Viamed Hospital, Santa Ángela de la Cruz, Seville, Spain
| | - Yolanda M Pacheco
- Immunology Laboratory, Institute of Biomedicine of Seville (IBiS), Immunology Service, Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Ave. Manuel Siurot s/n, 41013, Seville, Spain.
| |
Collapse
|
21
|
Benitez G, Shehadeh F, Mylona EK, Tran QL, Tsikala-Vafea M, Atalla E, Kaczynski M, Mylonakis E. Effect of Thymalfasin (Thymosin-α-1) on Reversing Lymphocytopenia among Patients with COVID-19. Int Immunopharmacol 2023:109831. [PMCID: PMC9902288 DOI: 10.1016/j.intimp.2023.109831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Introduction Thymosin-α-1 (Tα1) elevates lymphocyte counts among patients with COVID-19, but its effect on reversing lymphocytopenia is unknown. Methods 24 patients treated with Tα1 and 100 patients in the control arm were included in this analysis. The incidence rate of reversing lymphocytopenia, overall and stratified by baseline oxygen support, above the threshold for classification of lymphocytopenia (i.e., Total Lymphocyte Count (TLC) < 1.5 x 109/L) and severe lymphocytopenia (i.e., TLC < 1.0 x 109/L) within 3, 5, and 7 days of treatment initiation was calculated, along with incidence rate ratios (IRRs) and 95% confidence intervals (CIs). Results Compared with the standard of care, the rate of reversing lymphocytopenia (IRR: 2.38, 95% CI: 0.92 – 5.81) and severe lymphocytopenia (IRR: 1.57, 95% CI: 0.59 – 3.72), especially among patients with severe lymphocytopenia on high flow oxygen support (IRR: 3.64, 95% CI: 0.71 – 23.44), was greater for patients treated with Tα1 within 3 days of treatment initiation, although analyses were not significant. Conclusion Among patients with hypoxemia and lymphocytopenia, Tα1 may reverse lymphocytopenia and severe lymphocytopenia, particularly within 3 days of treatment initiation, faster than the standard of care.
Collapse
Affiliation(s)
- Gregorio Benitez
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Fadi Shehadeh
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,School of Electrical and Computer Engineering, National Technical University of Athens, Athens, Greece
| | - Evangelia K. Mylona
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Quynh-Lam Tran
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Maria Tsikala-Vafea
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Eleftheria Atalla
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Matthew Kaczynski
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Corresponding author at: Eleftherios Mylonakis 593 Eddy Street, POB, 3rd Floor, Suite 328/330, Providence, RI 02903, USA
| |
Collapse
|
22
|
Bulnes-Ramos Á, Pozo-Balado MM, Olivas-Martínez I, Garrido-Rodríguez V, Bernal-Blanco G, Suárez-Benjumea A, Álvarez-Ríos AI, Lozano C, González-Corvillo C, Suñer-Poblet M, González-Roncero FM, Sánchez B, Maldonado-Calzado I, Lara-Ruiz JM, Gonzalez-Escribano MF, Pacheco YM. Factors associated with the humoral response after three doses of COVID-19 vaccination in kidney transplant recipients. Front Immunol 2023; 14:1099079. [PMID: 36875099 PMCID: PMC9977967 DOI: 10.3389/fimmu.2023.1099079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Kidney transplant recipients showed a weak humoral response to the mRNA COVID-19 vaccine despite receiving three cumulative doses of the vaccine. New approaches are still needed to raise protective immunity conferred by the vaccine administration within this group of high-risk patients. Methods To analyze the humoral response and identify any predictive factors within these patients, we designed a prospective monocentric longitudinal study of Kidney transplant recipients (KTR) who received three doses of mRNA-1273 COVID-19 vaccine. Specific antibody levels were measured by chemiluminescence. Parameters related to clinical status such as kidney function, immunosuppressive therapy, inflammatory status and thymic function were analyzed as potential predictors of the humoral response. Results Seventy-four KTR and sixteen healthy controls were included. One month after the administration of the third dose of the COVID-19 vaccine, 64.8% of KTR showed a positive humoral response. As predictive factors of seroconversion and specific antibody titer, we found that immunosuppressive therapy, worse kidney function, higher inflammatory status and age were related to a lower response in KTR while immune cell counts, thymosin-a1 plasma concentration and thymic output were related to a higher humoral response. Furthermore, baseline thymosin-a1 concentration was independently associated with the seroconversion after three vaccine doses. Discussion In addition to the immunosuppression therapy, condition of kidney function and age before vaccination, specific immune factors could also be relevant in light of optimization of the COVID-19 vaccination protocol in KTR. Therefore, thymosin-a1, an immunomodulatory hormone, deserves further research as a potential adjuvant for the next vaccine boosters.
Collapse
Affiliation(s)
- Ángel Bulnes-Ramos
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - María Mar Pozo-Balado
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - Israel Olivas-Martínez
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - Vanesa Garrido-Rodríguez
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | | | | | | | - Carmen Lozano
- Microbiology Service, University Hospital Virgen del Rocío, Seville, Spain
| | | | - Marta Suñer-Poblet
- Nephrology Service, University Hospital Virgen del Rocío, Seville, Spain
| | | | - Berta Sánchez
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - Isabel Maldonado-Calzado
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - José Manuel Lara-Ruiz
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - María Francisca Gonzalez-Escribano
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| | - Yolanda María Pacheco
- Immunology Service, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital (HUVR)/CSIC/University of Seville, Seville, Spain
| |
Collapse
|
23
|
Shehadeh F, Benitez G, Mylona EK, Tran QL, Tsikala-Vafea M, Atalla E, Kaczynski M, Mylonakis E. A Pilot Trial of Thymalfasin (Thymosin-α-1) to Treat Hospitalized Patients With Hypoxemia and Lymphocytopenia Due to Coronavirus Disease 2019 Infection. J Infect Dis 2022; 227:226-235. [PMID: 36056913 PMCID: PMC9494344 DOI: 10.1093/infdis/jiac362] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/26/2022] [Accepted: 09/01/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Thymosin-α-1 (Tα1) may be a treatment option for coronavirus disease 2019 (COVID-19), but efficacy and safety data remain limited. METHODS Prospective, open-label, randomized trial assessing preliminary efficacy and safety of thymalfasin (synthetic form of Tα1), compared with the standard of care, among hospitalized patients with hypoxemia and lymphocytopenia due to COVID-19. RESULTS A total of 49 patients were included in this analysis. Compared with control patients, the incidence of clinical recovery was higher for treated patients with either baseline low-flow oxygen (subdistribution hazard ratio, 1.48 [95% confidence interval, .68-3.25]) or baseline high-flow oxygen (1.28 [.35-4.63]), although neither difference was significant. Among patients with baseline low-flow oxygen, treated patients, compared with control patients, had an average difference of 3.84 times more CD4+ T cells on day 5 than on day 1 (P = .01). Nine serious adverse events among treated patients were deemed not related to Tα1. CONCLUSIONS Tα1 increases CD4+ T-cell count among patients with baseline low-flow oxygen support faster than the standard of care and may have a role in the management of hospitalized patients with hypoxemia and lymphocytopenia due to COVID-19. CLINICAL TRIALS REGISTRATION NCT04487444.
Collapse
Affiliation(s)
| | | | - Evangelia K Mylona
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Quynh Lam Tran
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Maria Tsikala-Vafea
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eleftheria Atalla
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA,University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Matthew Kaczynski
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Correspondence to: Eleftherios Mylonakis 593 Eddy Street, POB, 3rd Floor, Suite 328/330, Providence, RI 02903, USA.
| |
Collapse
|
24
|
Genetic Associations and Differential mRNA Expression Levels of Host Genes Suggest a Viral Trigger for Endemic Pemphigus Foliaceus. Viruses 2022; 14:v14050879. [PMID: 35632621 PMCID: PMC9144834 DOI: 10.3390/v14050879] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
The long search for the environmental trigger of the endemic pemphigus foliaceus (EPF, fogo selvagem) has not yet resulted in any tangible findings. Here, we searched for genetic associations and the differential expression of host genes involved in early viral infections and innate antiviral defense. Genetic variants could alter the structure, expression sites, or levels of the gene products, impacting their functions. By analyzing 3063 variants of 166 candidate genes in 227 EPF patients and 194 controls, we found 12 variants within 11 genes associated with differential susceptibility (p < 0.005) to EPF. The products of genes TRIM5, TPCN2, EIF4E, EIF4E3, NUP37, NUP50, NUP88, TPR, USP15, IRF8, and JAK1 are involved in different mechanisms of viral control, for example, the regulation of viral entry into the host cell or recognition of viral nucleic acids and proteins. Only two of nine variants were also associated in an independent German cohort of sporadic PF (75 patients, 150 controls), aligning with our hypothesis that antiviral host genes play a major role in EPF due to a specific virus−human interaction in the endemic region. Moreover, CCL5, P4HB, and APOBEC3G mRNA levels were increased (p < 0.001) in CD4+ T lymphocytes of EPF patients. Because there is limited or no evidence that these genes are involved in autoimmunity, their crucial role in antiviral responses and the associations that we observed support the hypothesis of a viral trigger for EPF, presumably a still unnoticed flavivirus. This work opens new frontiers in searching for the trigger of EPF, with the potential to advance translational research that aims for disease prevention and treatment.
Collapse
|
25
|
Grønning AGB, Kacprowski T, Schéele C. MultiPep: a hierarchical deep learning approach for multi-label classification of peptide bioactivities. Biol Methods Protoc 2021; 6:bpab021. [PMID: 34909478 PMCID: PMC8665375 DOI: 10.1093/biomethods/bpab021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/28/2021] [Accepted: 11/17/2021] [Indexed: 11/14/2022] Open
Abstract
Peptide-based therapeutics are here to stay and will prosper in the future. A key step in identifying novel peptide-drugs is the determination of their bioactivities. Recent advances in peptidomics screening approaches hold promise as a strategy for identifying novel drug targets. However, these screenings typically generate an immense number of peptides and tools for ranking these peptides prior to planning functional studies are warranted. Whereas a couple of tools in the literature predict multiple classes, these are constructed using multiple binary classifiers. We here aimed to use an innovative deep learning approach to generate an improved peptide bioactivity classifier with capacity of distinguishing between multiple classes. We present MultiPep: a deep learning multi-label classifier that assigns peptides to zero or more of 20 bioactivity classes. We train and test MultiPep on data from several publically available databases. The same data are used for a hierarchical clustering, whose dendrogram shapes the architecture of MultiPep. We test a new loss function that combines a customized version of Matthews correlation coefficient with binary cross entropy (BCE), and show that this is better than using class-weighted BCE as loss function. Further, we show that MultiPep surpasses state-of-the-art peptide bioactivity classifiers and that it predicts known and novel bioactivities of FDA-approved therapeutic peptides. In conclusion, we present innovative machine learning techniques used to produce a peptide prediction tool to aid peptide-based therapy development and hypothesis generation.
Collapse
Affiliation(s)
- Alexander G B Grønning
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, TU Braunschweig and Hannover Medical School, 38106 Braunschweig, Germany.,Braunschweig Integrated Centre for Systems Biology (BRICS), 38106 Braunschweig, Germany
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
26
|
Abstract
Sepsis is expected to have a substantial impact on public health and cost as its prevalence increases. Factors contributing to increased prevalence include a progressively aging population, advances in the use of immunomodulatory agents to treat a rising number of diseases, and immune-suppressing therapies in organ transplant recipients and cancer patients. It is now recognized that sepsis is associated with profound and sustained immunosuppression, which has been implicated as a predisposing factor in the increased susceptibility of patients to secondary infections and mortality. In this review, we discuss mechanisms of sepsis-induced immunosuppression and biomarkers that identify a state of impaired immunity. We also highlight immune-enhancing strategies that have been evaluated in patients with sepsis, as well as therapeutics under current investigation. Finally, we describe future challenges and the need for a new treatment paradigm, integrating predictive enrichment with patient factors that may guide the future selection of tailored immunotherapy. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Lisa K Torres
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, New York, NY, USA;
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands;
| | - Tom van der Poll
- Center of Experimental and Molecular Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands;
| |
Collapse
|
27
|
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
|
28
|
Aslam MS, Zaidi SZJ, Toor RH, Gull I, Iqbal MM, Abbas Z, Tipu I, Ahmed A, Athar MA, Harito C, Hassan SU. Interferon α2-Thymosin α1 Fusion Protein (IFNα2-Tα1): A Genetically Engineered Fusion Protein with Enhanced Anticancer and Antiviral Effect. MATERIALS 2021; 14:ma14123318. [PMID: 34203928 PMCID: PMC8232609 DOI: 10.3390/ma14123318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/09/2021] [Accepted: 05/09/2021] [Indexed: 12/30/2022]
Abstract
Human interferon α2 (IFNα2) and thymosin α1 (Tα1) are therapeutic proteins used for the treatment of viral infections and different types of cancer. Both IFNα2 and Tα1 show a synergic effect in their activities when used in combination. Furthermore, the therapeutic fusion proteins produced through the genetic fusion of two genes can exhibit several therapeutic functions in one molecule. In this study, we determined the anticancer and antiviral effect of human interferon α2-thymosin α1 fusion protein (IFNα2-Tα1) produced in our laboratory for the first time. The cytotoxic and genotoxic effect of IFNα2-Tα1 was evaluated in HepG2 and MDA-MB-231 cells. The in vitro assays confirmed that IFNα2-Tα1 inhibited the growth of cells more effectively than IFNα2 alone and showed an elevated genotoxic effect. The expression of proapoptotic genes was also significantly enhanced in IFNα2-Tα1-treated cells compared to IFNα2-treated cells. Furthermore, the HCV RNA level was significantly reduced in IFNα2-Tα1-treated HCV-infected Huh7 cells compared to IFNα2-treated cells. The quantitative PCR analysis showed that the expression of various genes, the products of which inhibit HCV replication, was significantly enhanced in IFNα2-Tα1-treated cells compared to IFNα2-treated cells. Our findings demonstrate that IFNα2-Tα1 is more effective than single IFNα2 as an anticancer and antiviral agent.
Collapse
Affiliation(s)
- Muhammad Shahbaz Aslam
- Institute of Biochemistry and Biotechnology, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (I.G.); (M.M.I.); (M.A.A.)
- Correspondence: (M.S.A.); (S.Z.J.Z.); (S.-u.H.)
| | - Syed Zohaib Javaid Zaidi
- Institute of Chemical Engineering and Technology, University of the Punjab, Lahore 54590, Pakistan
- Correspondence: (M.S.A.); (S.Z.J.Z.); (S.-u.H.)
| | - Rabail Hassan Toor
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (R.H.T.); (A.A.)
| | - Iram Gull
- Institute of Biochemistry and Biotechnology, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (I.G.); (M.M.I.); (M.A.A.)
| | - Muhammad Mudassir Iqbal
- Institute of Biochemistry and Biotechnology, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (I.G.); (M.M.I.); (M.A.A.)
| | - Zaigham Abbas
- Department of Microbiology & Molecular Genetics, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan;
| | - Imran Tipu
- Department of Life Sciences, University of Management & Technology, Lahore 54770, Pakistan;
| | - Aftab Ahmed
- School of Biological Sciences, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (R.H.T.); (A.A.)
| | - Muhammad Amin Athar
- Institute of Biochemistry and Biotechnology, University of the Punjab, Quaid-i-Azam Campus, Lahore 54590, Pakistan; (I.G.); (M.M.I.); (M.A.A.)
| | - Christian Harito
- Industrial Engineering Department, Faculty of Engineering, Bina Nusantara University, Jakarta 11480, Indonesia;
| | - Sammer-ul Hassan
- Bioengineering Research Group, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton SO17 1BJ, UK
- Correspondence: (M.S.A.); (S.Z.J.Z.); (S.-u.H.)
| |
Collapse
|
29
|
Huang C, Fei L, Xu W, Li W, Xie X, Li Q, Chen L. Efficacy Evaluation of Thymosin Alpha 1 in Non-severe Patients With COVID-19: A Retrospective Cohort Study Based on Propensity Score Matching. Front Med (Lausanne) 2021; 8:664776. [PMID: 33968969 PMCID: PMC8102900 DOI: 10.3389/fmed.2021.664776] [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] [Received: 02/06/2021] [Accepted: 03/16/2021] [Indexed: 12/15/2022] Open
Abstract
Objective: Thymosin alpha 1 (Thymosin-α1) is a potential treatment for patients with COVID-19. We aimed to determine the effect of Thymosin-α1 in non-severe patients with COVID-19. Methods: We retrospectively enrolled 1,388 non-severe patients with COVID-19. The primary and secondary clinical outcomes were evaluated with comparisons between patients treated with or without Thymosin-α1 therapy. Results: Among 1,388 enrolled patients, 232 patients (16.7%) received both Thymosin-α1 therapy and standard therapy (Thymosin-α1 group), and 1,156 patients (83.3%) received standard therapy (control group). After propensity score matching (1:1 ratio), baseline characteristics were well-balanced between the Thymosin-α1 group and control group. The proportion of patients that progressed to severe COVID-19 is 2.17% for the Thymosin-α1 group and 2.71% for the control group (p = 0.736). The COVID-19-related mortality is 0.54% for the Thymosin-α1 group and 0 for the control group (p = 0.317). Compared with the control group, the Thymosin-α1 group had significantly shorter SARS-CoV-2 RNA shedding duration (13 vs. 16 days, p = 0.025) and hospital stay (14 vs. 18 days, p < 0.001). No statistically significant difference was found between the Thymosin-α1 group and control group in duration of symptoms (median, 4 vs. 3 days, p = 0.843) and antibiotic utilization rate (14.1% vs. 15.2%, p = 0.768). Conclusion: For non-severe patients with COVID-19, Thymosin-α1 can shorten viral RNA shedding duration and hospital stay but did not prevent COVID-19 progression and reduce COVID-19-related mortality rate.
Collapse
Affiliation(s)
- ChenLu Huang
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ling Fei
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei Xu
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - WeiXia Li
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - XuDong Xie
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Qiang Li
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Liang Chen
- Department of Liver Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Liu T, Liu S, Li T, Zhang J. Thymosin a1 use is not associated with reduced COVID-19 mortality. Am J Emerg Med 2021; 53:252-253. [PMID: 33896653 PMCID: PMC8053599 DOI: 10.1016/j.ajem.2021.04.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Tao Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang 550001, China
| | - Shengdong Liu
- Department of Critical Care Medicine, the First Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang 550001, China
| | - Tao Li
- Cardiology, the Second Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang 550001, China
| | - Jingjing Zhang
- Renal medicine, the First Affiliated Hospital of Guizhou University of Chinese Medicine, Guiyang 550001, China.
| |
Collapse
|
31
|
Giuliani C, Verrocchio S, Verginelli F, Bucci I, Grassadonia A, Napolitano G. Hormonal Regulation of the MHC Class I Gene in Thyroid Cells: Role of the Promoter "Tissue-Specific" Region. Front Endocrinol (Lausanne) 2021; 12:749609. [PMID: 34938270 PMCID: PMC8685237 DOI: 10.3389/fendo.2021.749609] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/15/2021] [Indexed: 11/18/2022] Open
Abstract
In previous studies we have demonstrated that the expression of the Major Histocompatibility Complex (MHC) class I gene in thyrocytes is controlled by several hormones, growth factors, and drugs. These substances mainly act on two regions of the MHC class I promoter a "tissue-specific" region (-800 to -676 bp) and a "hormone/cytokines-sensitive" region (-500 to -68 bp). In a previous study, we have shown that the role of the "tissue-specific" region in the MHC class I gene expression is dominant compared to that of the "hormone/cytokines-sensitive" region. In the present report we further investigate the dominant role of the "tissue-specific" region evaluating the effect of thyroid stimulating hormone (TSH), methimazole (MMI), phenylmethimazole (C10), glucose and thymosin-α1. By performing experiments of electrophoretic mobility shift assays (EMSAs) we show that TSH, MMI and C10, which inhibit MHC class I expression, act on the "tissue-specific" region increasing the formation of a silencer complex. Glucose and thymosin-α1, which stimulate MHC class I expression, act decreasing the formation of this complex. We further show that the silencer complex is formed by two distinct members of the transcription factors families activator protein-1 (AP-1) and nuclear factor-kB (NF-kB), c-jun and p65, respectively. These observations are important in order to understand the regulation of MHC class I gene expression in thyroid cells and its involvement in the development of thyroid autoimmunity.
Collapse
Affiliation(s)
- Cesidio Giuliani
- Unit of Endocrinology, Department of Medicine and Sciences of Aging, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- *Correspondence: Cesidio Giuliani,
| | - Sara Verrocchio
- Unit of Endocrinology, Department of Medicine and Sciences of Aging, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Fabio Verginelli
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Ines Bucci
- Unit of Endocrinology, Department of Medicine and Sciences of Aging, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Antonino Grassadonia
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Department of Oral, Medical and Biotechnological Science, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Giorgio Napolitano
- Unit of Endocrinology, Department of Medicine and Sciences of Aging, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Centre for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| |
Collapse
|