Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
BMJ Open ; 14(3): e075642, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38458816

ABSTRACT

INTRODUCTION: The PRaG regimen, which consists of hypofractionated radiotherapy combined with a programmed cell death-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitor and granulocyte-macrophage colony stimulating factor (GM-CSF), has been demonstrated to have a survival benefit in patients with advanced solid tumours who have failed at least two lines of treatment. Nonetheless, lymphopenia poses an impediment to the enduring efficacy of PD-1/PD-L1 inhibitor therapy. Adequate lymphocyte reserves are essential for the efficacy of immunotherapy. Coupling the PRaG regimen with immunomodulatory agents that augment the number and functionality of lymphocytes may yield further survival benefits in this cohort of patients. OBJECTIVE: The aim of this study is to investigate the effectiveness and safety of a meticulously thymalfasin-controlled PRaG regimen in patients with advanced and chemotherapy-resistant solid tumours. METHODS AND ANALYSIS: The study has a prospective, single-arm, open-label, multicentre design and aims to recruit up to 60 patients with histologically confirmed advanced solid tumours that have relapsed or metastasised. All eligible patients will receive a minimum of two cycles of the PRaG regimen comprising thymalfasin followed by maintenance treatment with a PD-1/PD-L1 inhibitor and thymalfasin for 1 year or until disease progression. Patients will be monitored according to the predetermined protocol for a year or until disease progression after initiation of radiotherapy. ETHICS AND DISSEMINATION: The study protocol was approved by the Ethics Committee of the Second Affiliated Hospital of Soochow University, on 25 November 2022 (JD-LK-2022-151-01) and all other participating hospitals. Findings will be disseminated through national and international conferences. We also plan to publish our findings in high-impact peer-reviewed journal. TRIAL REGISTRATION NUMBER: NCT05790447.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Thymalfasin/therapeutic use , Prospective Studies , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/therapeutic use , Neoplasms/drug therapy , Disease Progression , Antineoplastic Combined Chemotherapy Protocols , Multicenter Studies as Topic
2.
Altern Ther Health Med ; 30(1): 6-12, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38308608

ABSTRACT

Objective: This study aims to assess the safety and efficacy of Thymosin Alpha 1 (Tα1) through a comprehensive narrative review of clinical studies involving over 11 000 human subjects in more than 30 trials. The focus was on Tα1's application in COVID-19, autoimmune conditions, and cancer treatment, with implications for future considerations. Methods: We systematically searched articles relevant to critical studies on COVID-19, infectious diseases, cancer, and autoimmune diseases indexed on Pubmed, Google Scholar, and Cochrane Library. Our focus was on evaluating the safety and efficacy of Tα1 in human subjects. Clinical trials conducted worldwide involving diverse populations were analyzed to assess the safety and effectiveness of Tα1. The review examines explicit outcomes in over 11 000 human subjects, emphasizing its role in addressing COVID-19, autoimmune conditions, and cancer treatment. Results: Contrary to the FDA's restriction on Tα1 and 21 additional peptides in 2023, our analysis reveals consistent evidence of Tα1's safety and efficacy. The peptide has demonstrated significant effectiveness in treating various conditions, including COVID-19, autoimmune disorders, and cancer. This review summarizes conclusions drawn from a comprehensive examination of clinical trials worldwide. Conclusions: Based on substantial evidence from clinical trials, Tα1 emerges as a well-tolerated and effective immune modulator. The FDA>s restriction appears unfounded, as Tα1 has shown safety and efficacy beyond the initially specified conditions. Urgent attention and intervention are warranted to ensure the continued availability of this life-saving peptide through prescription. Therefore, it is recommended that the FDA permits 503A compounding pharmacies to compound Tα1, considering its potential to treat a variety of conditions effectively.


Subject(s)
Autoimmune Diseases , COVID-19 , Neoplasms , Thymosin , Humans , Thymalfasin/therapeutic use , Thymosin/therapeutic use , Autoimmune Diseases/drug therapy , Neoplasms/drug therapy
3.
BMC Infect Dis ; 24(1): 97, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38233816

ABSTRACT

BACKGROUND: Immunological nonresponders (INRs) living with HIV are at increased risk of co-infection and multiple tumors, with no effective strategy currently available to restore their T-cell immune response. This study aimed to explore the safety and efficacy of thymosin α1 in reconstituting the immune response in INRs. METHODS: INRs with CD4 + T cell counts between 100 and 350 cells/µL were enrolled and received two-staged 1.6 mg thymosin α1 subcutaneous injections for 24 weeks (daily in the first 2 weeks and biweekly in the subsequent 22 weeks) while continuing antiretroviral therapy. T cell counts and subsets, the expression of PD-1 and TIM-3 on T cells, and signal joint T cell receptor excision circles (sjTREC) at week 24 were evaluated as endpoints. RESULTS: Twenty three INRs were screened for eligibility, and 20 received treatment. The majority were male (19/20), with a median age of 48.1 years (interquartile range: 40.5-57.0) and had received antiretroviral therapy for 5.0 (3.0, 7.3) years. Multiple comparisons indicated that CD4 + T cell count and sjTREC increased after initiation of treatment, although no significant differences were observed at week 24 compared to baseline. Greatly, levels of CD4 + T cell proportion (17.2% vs. 29.1%, P < 0.001), naïve CD4 + and CD8 + T cell proportion (17.2% vs. 41.1%, P < 0.001; 13.8% vs. 26.6%, P = 0.008) significantly increased. Meanwhile, the proportion of CD4 + central memory T cells of HIV latent hosts (42.7% vs. 10.3%, P < 0.001) significantly decreased. Moreover, the expression of PD-1 on CD4 + T cells (14.1% vs. 6.5%, P < 0.001) and CD8 + T cells (8.5% vs. 4.1%, P < 0.001) decreased, but the expression of TIM-3 on T cellsremained unaltered at week 24. No severe adverse events were reported and HIV viral loads kept stable throughout the study. CONCLUSIONS: Thymosin α1 enhance CD4 + T cell count and thymic output albeit as a trend rather than an endpoint. Importantly, it improves immunosenescence and decreases immune exhaustion, warranting further investigation. TRIAL REGISTRATION: This single-arm prospective study was registered with ClinicalTrials.gov (NCT04963712) on July 15, 2021.


Subject(s)
HIV Infections , Hepatitis A Virus Cellular Receptor 2 , Humans , Male , Female , Adult , Middle Aged , Thymalfasin/therapeutic use , Programmed Cell Death 1 Receptor , Prospective Studies , CD4-Positive T-Lymphocytes , HIV Infections/drug therapy , CD4 Lymphocyte Count , Immunity
4.
Inflammopharmacology ; 31(6): 3317-3325, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37845598

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.


Subject(s)
COVID-19 , Humans , Thymalfasin/therapeutic use , Respiration, Artificial
5.
Int Immunopharmacol ; 124(Pt B): 110983, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37769533

ABSTRACT

BACKGROUND: The Coronavirus disease-19 (COVID-19) pandemic has posed a serious threat to global health. Thymosin α1 (Tα1) was considered to be applied in COVID-19 therapy. However, the data remains limited. METHODS: Participants with or without Tα1 treatment were recruited. Single cell RNA-sequencing (scRNA-seq) and T cell receptor-sequencing (TCR-seq) of the peripheral blood mononuclear cell (PBMC) samples were done to analyze immune features. The differential expression analysis and functional enrichment analysis were performed to explore the mechanism of Tα1 therapy. RESULTS: 33 symptomatic SARS-CoV-2-infected individuals (COV) and 11 healthy controls (HC) were enrolled in this study. The proportion of CD3+ KLRD1+ NKT, TBX21+ CD8+ NKT was observed to increase in COVID-19 patients with Tα1 treatment (COVT) than those without Tα1 (COV) (p = 0.024; p = 0.010). These two clusters were also significantly higher in Health controls with Tα1 treatment (HCT) than those without Tα1 (HC) (p = 0.016; p = 0.031). Besides, a series of genes and pathways related to immune responses were significantly higher enriched in Tα1 groups TBX21+ CD8+ NKT, such as KLRB1, PRF1, natural killer cell-mediated cytotoxicity pathway, chemokine signaling pathway, JAK-STAT signaling pathway. The increased TRBV9-TRBJ1-1 pair existed in both HCs and COVID-19 patients after Tα1 treatment. 1389 common complementarity determining region 3 nucleotides (CDR 3 nt) were found in COV and HC, while 0 CDR 3 nt was common in COVT and HCT. CONCLUSIONS: Tα1 increased CD3+ KLRD1+ NKT, TBX21+ CD8+ NKT cell proportion and stimulated the diversity of TCR clones in COVT and HCT. And Tα1 could regulate the expression of genes associated with NKT activation or cytotoxicity to promote NKT cells. These data support the use of Tα1 in COVID-19 patients.


Subject(s)
COVID-19 , Thymosin , Humans , Thymalfasin/therapeutic use , Thymosin/genetics , Thymosin/metabolism , Leukocytes, Mononuclear/metabolism , SARS-CoV-2/metabolism , Receptors, Antigen, T-Cell/genetics
6.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37295817

ABSTRACT

BACKGROUND: Advanced or metastatic prostate cancer (PCa) is still an incurable malignancy with high lethality and a poor prognosis. Despite the remarkable success of immunotherapy against many types of cancer, most patients with PCa receive minimal benefit from current immunotherapeutic strategies, because PCa is an immune cold tumor with scarce T-cell infiltration in the tumor microenvironment. The aim of this study was to develop an effective immunotherapeutic approach for immune cold PCa tumors. METHODS: The therapeutic efficacy of androgen deprivation therapy (ADT) and zoledronic acid (ZA) plus thymosin α1 (Tα1) therapy was analyzed retrospectively in patients with advanced or metastatic PCa. The effects and mechanisms by which ZA and Tα1 regulated the immune functions of PCa cells and immune cells were evaluated by a PCa allograft mouse model, flow cytometric analysis, immunohistochemical and immunofluorescence staining assays, and PCR, ELISA, and Western blot analyses. RESULTS: In this study, clinical retrospective analysis revealed that ADT combined with ZA plus Tα1 improved the therapeutic outcomes of patients with PCa, which might be associated with an enhanced frequency of T cells. ZA and Tα1 treatment synergistically inhibited the growth of androgen-independent PCa allograft tumors, with increased infiltration of tumor-specific cytotoxic CD8+ T cells and enhanced tumor inflammation. Functionally, ZA and Tα1 treatment relieved immunosuppression in PCa cells, stimulated pro-inflammatory macrophages, and enhanced the cytotoxic function of T cells. Mechanistically, ZA plus Tα1 therapy blocked the MyD88/NF-κB pathway in PCa cells but activated this signaling in macrophages and T cells, altering the tumor immune landscape to suppress PCa progression. CONCLUSIONS: These findings uncover a previously undefined role for ZA and Tα1 in inhibiting the disease progression of immune cold PCa tumors by enhancing antitumor immunity and pave the way for the application of ZA plus Tα1 therapy as an immunotherapeutic strategy for treating patients with immunologically unresponsive PCa.


Subject(s)
Prostatic Neoplasms , Humans , Male , Animals , Mice , Zoledronic Acid/pharmacology , Zoledronic Acid/therapeutic use , Thymalfasin/pharmacology , Thymalfasin/therapeutic use , Prostatic Neoplasms/pathology , Retrospective Studies , T-Lymphocytes, Cytotoxic/metabolism , CD8-Positive T-Lymphocytes/metabolism , Androgen Antagonists/therapeutic use , Androgens/therapeutic use , Inflammation , Tumor Microenvironment
7.
Int Immunopharmacol ; 117: 109952, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36871535

ABSTRACT

Thymosin alpha 1 (Tα1) is a highly conserved 28 amino-acid peptide naturally occurring in the thymus and plays critical roles in T cell maturity and differentiation. Its synthetic form, thymalfasin, has been approved by various regulatory agencies in the treatment of hepatitis B viral infection and as an enhancer of vaccine response in immune-compromised populations. In China, it has also widely utilized in patients with cancer and severe infections, as well as the emergency use during (Severe Acute Respiratory Syndrome)SARS and COVID-19 pandemic as an immune-regulator. Recent studies showed that Tα1 could significantly improve overall survival (OS) in patients with surgically resectable non-small cell lung cancer (NSCLC) and liver cancers in the adjuvant setting. For patients with locally advanced, unresectable NSCLC, Tα1 could significantly reduce chemoradiation-induced lymphopenia, pneumonia, and trending improvement of OS. Preclinical evidence are emerging to demonstrate that Tα1 may augment efficacy of cancer chemotherapy by reversing efferocytosis-induced M2 polarization of macrophages via activation of a TLR7/SHIP1 axis and enhancing anti-tumor immunity by turning "cold-tumors" to "hot-tumors"; a protective role in reducing colitis caused by immune check-point inhibitors (ICIs). Potential enhancement of ICIs' clinical efficacies has also been indicated. ICIs have transformed ways treating patients with cancer but limitations such as relatively low response rates and certain safety issues remains. Given the roles of Tα1 in regulating cellular immunities and exceptional safety profiles demonstrated in decades clinical uses, we believe that it is plausible to explore implications of Tα1 the immune-oncology setting by combining with ICI-based therapeutic strategies. Background Activities of Tα1. Tα1 is a biological response modifier which activates various cells in the immune system [1-3]. Tα1 is therefore expected to have clinical benefits in disorders where immune responses are impaired or ineffective. These disorders include acute and chronic infections, cancers, and vaccine non-responsiveness. In severe sepsis, for example, sepsis-induced immunosuppression is increasingly recognized as the overriding immune dysfunction in these vulnerable patients [4] and there is now agreement that many patients with severe sepsis survive the first critical hours of the syndrome but eventually die later due to patients' immunosuppression which make the system difficulty to fight the primary bacterial infection, decreased resistance to secondary nosocomial infections, and reactivation of viral infections [5]. Tα1 has been shown to restore immune functions and help to reduce mortality in patients with severe sepsis.


Subject(s)
COVID-19 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Sepsis , Thymosin , Humans , Thymalfasin/therapeutic use , Thymosin/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Pandemics , COVID-19/therapy , Lung Neoplasms/drug therapy , Sepsis/drug therapy
8.
Int Immunopharmacol ; 117: 109949, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36881979

ABSTRACT

The recent COVID-19 pandemic has catalyzed the attention of the scientific community to the long-standing issue of lower respiratory tract infections. The myriad of airborne bacterial, viral and fungal agents to which humans are constantly exposed represents a constant threat to susceptible individuals and bears the potential to reach a catastrophic scale when the ease of inter-individual transmission couples with a severe pathogenicity. While we might be past the threat of COVID-19, the risk of future outbreaks of respiratory infections is tangible and argues for a comprehensive assessment of the pathogenic mechanisms shared by airborne pathogens. On this regard, it is clear that the immune system play a major role in dictating the clinical course of the infection. A balanced immune response is required not only to disarm the pathogens, but also to prevent collateral tissue damage, thus moving at the interface between resistance to infection and tolerance. Thymosin alpha1 (Tα1), an endogenous thymic peptide, is increasingly being recognized for its ability to work as an immunoregulatory molecule able to balance a derailed immune response, working as immune stimulatory or immune suppressive in a context-dependent manner. In this review, we will take advantage from the recent work on the COVID-19 pandemic to reassess the role of Tα1 as a potential therapeutic molecule in lung infections caused by either defective or exaggerated immune responses. The elucidation of the immune regulatory mechanisms of Tα1 might open a new window of opportunity for the clinical translation of this enigmatic molecule and a potential new weapon in our arsenal against lung infections.


Subject(s)
COVID-19 , Thymosin , Humans , Thymalfasin/therapeutic use , Thymosin/therapeutic use , Pandemics , Lung
9.
Int Immunopharmacol ; 117: 109950, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36881981

ABSTRACT

PURPOSE: Patients with end-stage renal disease (ESRD) on hemodialysis (HD) are considered particularly susceptible to infection with SARS-CoV2 on the basis of the immunodeficiency associated with advanced age, comorbidity burden, medication use, and need for frequent visits to dialysis clinics. In prior studies, thymalfasin (thymosin alpha 1, Ta1) has been shown to enhance antibody response to influenza vaccine and reduce influenza infection in geriatric populations, including hemodialysis patients, when used as an adjunct to influenza vaccine. Early in the COVID-19 pandemic we speculated that administration of Ta1 to HD patients would result in reduced rate and severity of COVID-19 infection. We also hypothesized that HD patients treated with Ta1 who did become infected with COVID-19 would have a milder course of infection in terms of hospitalization rates, requirement for and length of ICU stays, requirement for mechanical ventilation, and survival. Further, we proposed that patients who avoided COVID-19 infection during the study would have decreased non-COVID-19 infections and hospitalizations compared to controls. PROCEDURES: The study launched in January 2021 and, as of July 1, 2022, 254 ESRD/ HD patients from five dialysis centers in Kansas City, MO have been screened. Of these, 194 patients have been randomized 1:1 to either Group A (1.6 mg Ta1 given subcutaneously twice weekly for 8 weeks), or Group B (control group not receiving Ta1). After the 8-week treatment period, subjects were followed for an additional 4 months and monitored for safety and efficacy. A data safely monitoring board reviewed all reported adverse effects and commented on study progress. RESULTS: To date, only 3 deaths have occurred in subjects treated with Ta1 (Group A), compared to 7 in the control (Group B). There have been 12 COVID-19 related serious adverse effects (SAEs; 5 in Group A, and 7 in Group B). The majority of patients have received a COVID-19 vaccine (91 patients in group A, and 76 patients in Group B) at various times throughout the study. Nearing completion of the study, blood samples have been collected and antibody responses to COVID-19 will be analyzed along with safety and efficacy endpoints when all subjects have completed the study.


Subject(s)
COVID-19 , Influenza Vaccines , Kidney Failure, Chronic , Humans , Aged , COVID-19/epidemiology , Thymalfasin/therapeutic use , SARS-CoV-2/genetics , COVID-19 Vaccines , Pandemics/prevention & control , RNA, Viral , Pilot Projects , Renal Dialysis , Kidney Failure, Chronic/therapy , Morbidity
10.
Int Immunopharmacol ; 118: 110055, 2023 May.
Article in English | MEDLINE | ID: mdl-36989892

ABSTRACT

The complex alterations of the immune system and the immune-mediated multiorgan injury plays a key role in host response to SARS-CoV-2 infection and in the pathogenesis of COVID-19, being also associated with adverse outcomes. Thymosin alpha 1 (Tα1) is one of the molecules used in the treatment of COVID-19, as it is known to restore the homeostasis of the immune system during infections and cancer. The use of Tα1 in COVID-19 patients had been widely used in China and in COVID-19 patients, it has been shown to decrease hospitalization rate, especially in those with greater disease severity, and reduce mortality by restoring lymphocytopenia and more specifically, depleted T cells. Persistent dysregulation with depletion of naive B and T cell subpopulations and expansion of memory T cells suggest a chronic stimulation of the immune response in individuals with post-acute sequelae of SARS-CoV-2 infection (PASC). Our data obtained from an ex vivo study, showed that in PASC individuals with a chronically altered immune response, Tα1 improve the restoration of an appropriate response, most evident in those with more severe illness and who need respiratory support during acute phase, and in those with specific systemic and psychiatric symptoms of PASC, confirming Tα1 treatment being more effective in compromised patients. The results obtained, along with promising reports on recent trials on Tα1 administration in patients with COVID-19, offer new insights into intervention also for those patients with long-lasting inflammation with post-infectious symptoms, some of which have a delayed onset.


Subject(s)
COVID-19 , Thymosin , Humans , Thymalfasin/therapeutic use , Post-Acute COVID-19 Syndrome , SARS-CoV-2 , Lymphocytes , Homeostasis , Thymosin/therapeutic use
11.
Int Immunopharmacol ; 117: 109996, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36933449

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.


Subject(s)
COVID-19 , Thymosin , Humans , Thymalfasin/therapeutic use , Leukocytes, Mononuclear/metabolism , SARS-CoV-2/metabolism , Cytokines/metabolism , Inflammation/drug therapy , Thymosin/pharmacology , Thymosin/therapeutic use
12.
Int Immunopharmacol ; 116: 109829, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36758296

ABSTRACT

BACKGROUND: Peritoneal metastases of colorectal carcinoma origin (PM-CRC) are treated by cytoreductive surgery and heated intraperitoneal chemotherapy (HIPEC). However, the majority of patients recur, calling for novel treatments. We explored the immunogenic changes induced by HIPEC and the possibility to use thymosin α1 (Tα1) as an immune-stimulatory agent. METHODS: We used an experimental murine model of PM-CRC combined with mitomycin (MMC)-based HIPEC. We determined immune cell infiltration into tumor metastases after HIPEC administration by means of immunohistochemistry, and determined immunogenic cell death signals in tumor cells by real-time polymerase chain reaction. RESULTS: Mice with PM-CRC treated by HIPEC had increased overall survival (OS) compared to sham-treated mice (median OS 22.8 vs 18.9 days, respectively; P < 0.001). HIPEC induced increased infiltration of CD4+, CD8+, CD68 + and CD20 + cells into omental and visceral metastases at a magnitude of 40-100 %. We searched for potential immune signals induced by HIPEC by determining its effects on known immunogenic cell death proteins (heat-shock protein [HSP]-70, HSP-90 and calreticulin). HIPEC significantly increased HSP-90 mRNA expression (2.37 ± 1.5 vs 1-fold change, P < 0.05). The OS of Tα1 treated mice significantly improved compared to HIPEC-treated mice (16.3 ± 0.8 vs 14.1 ± 0.6 days, respectively, P = 0.02) and vs sham (11.8 ± 0.8 days, P = 0.007). CONCLUSIONS: HIPEC induced immunogenic changes that led to increased immune cell infiltration. These changes were further augmented by Tα1 treatment. Future studies aimed at optimizing Tα1 treatment should focus upon the immune response it evokes.


Subject(s)
Colorectal Neoplasms , Hyperthermia, Induced , Peritoneal Neoplasms , Animals , Mice , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Thymalfasin/therapeutic use , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Neoplasm Recurrence, Local/drug therapy , Mitomycin/therapeutic use , Combined Modality Therapy , Survival Rate
13.
Int Immunopharmacol ; 117: 109744, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36812669

ABSTRACT

Thymosin α-1 (Tα-1) is an immunomodulating polypeptide of 28 amino acids, which was the first peptide isolated from thymic tissue and has been widely used for the treatment of viral infections, immunodeficiencies, and especially malignancies. Tα-1 stimulates both innate and adaptive immune responses, and its regulation of innate immune cells and adaptive immune cells varies under different disease conditions. Pleiotropic regulation of immune cells by Tα-1 depends on activation of Toll-like receptors and its downstream signaling pathways in various immune microenvironments. For treatment of malignancies, the combination of Tα-1 and chemotherapy has a strong synergistic effect by enhancing the anti-tumor immune response. On the basis of the pleiotropic effect of Tα-1 on immune cells and the promising results of preclinical studies, Tα-1 may be a favorable immunomodulator to enhance the curative effect and decrease immune-related adverse events of immune checkpoint inhibitors to develop novel cancer therapies.


Subject(s)
Neoplasms , Thymosin , Humans , Thymalfasin/therapeutic use , Thymalfasin/pharmacology , Thymosin/therapeutic use , Adjuvants, Immunologic/therapeutic use , Neoplasms/drug therapy , Immunity , Tumor Microenvironment
15.
Int Immunopharmacol ; 114: 109584, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36527881

ABSTRACT

OBJECTIVE: Thymosin alpha1 (Ta1) is widely used to treat patients with coronavirus disease 2019 (COVID-19), however, its effect remains unclear. This systematic review and meta-analysis aimed to evaluate the effect of Ta1 as a COVID-19 therapy. METHODS: PubMed, EMBASE, the Cochrane library, Web of Science, and the reference lists of relevant articles were searched to identify eligible studies. Assessment of heterogeneity was done using the I-squared (I2) test and random/fixed effect analysis was done to determine the risk ratio (RR). We polled the data related to mortality mainly by using Review Manager 5.4. Predefined subgroup analyses and sensitivity analyses were also performed. RESULTS: A total of 9 studies were included, on a total of 5352 (Ta1 = 1152, control = 4200) patient outcomes. Meta-analysis results indicated that Ta1 therapy had no statistically significant effect on mortality [RR 1.03 (0.60, 1.75), p = 0.92, I2 = 90 %]. Subgroup analyses demonstrated that the beneficial effect in mortality was associated with mean age>60 years in the Tα1 group [RR 0.68 (0.58, 0.78), p < 0.0000.1, I2 = 0 %], the proportion of female ≤ 40 % in the Tα1 group [RR 0.67 (0.58, 0.77), p < 0.0000.1, I2 = 0 %], and severe/critical COVID-19 patients [RR 0.66 (0.57, 0.76), p < 0.0000.1, I2 = 0 %]. Sensitivity analysis further demonstrated the results to be robust. CONCLUSIONS: The results of this meta-analysis do not support the use of Ta1 in hospitalized adult COVID-19 patients.


Subject(s)
COVID-19 , Thymosin , Humans , Adult , Female , Middle Aged , Thymalfasin/therapeutic use , Thymosin/therapeutic use
16.
J Infect Dis ; 227(2): 226-235, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36056913

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.


Subject(s)
COVID-19 , Lymphopenia , Thymosin , Humans , Thymalfasin/therapeutic use , Thymosin/therapeutic use , COVID-19/complications , Pilot Projects , Prospective Studies , Hypoxia/therapy , Hypoxia/drug therapy , Oxygen
17.
Altern Ther Health Med ; 28(7): 146-152, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35951068

ABSTRACT

Context: Septic shock (SS) can pose a high risk of death if rescue efforts in an emergency room aren't started in a timely manner. Thus, rapid and efficient treatment is of great significance to the SS patients' survival. T-α1 can enhance the cellular immune function of patients, and blood purification (BP) can improve the hemodynamics of SS patients by clearing inflammatory mediators in the blood. Objective: The study intended to explore the effects of Thymosin α1 (T-α1) plus blood purification (BP) on SS patients under the emergency green channel (GC), a fast and efficient service system that hospitals provide for acutely and critically ill patients. Design: The research team designed a randomized controlled study. Setting: The study took place in the Emergency Department at the Second Affiliated Hospital of Xi'an Jiaotong University in Xi'an, Shaanxi, China. Participants: Participants were 86 SS patients who came to the hospital for treatment between June 2019 and January 2021. Intervention: The research team numbered the patients in sequence according to the admission time of the patients, and then randomly numbered them by the computer, and assigned participants to an intervention or a control group, with 43 participants in the intervention group receiving T-α1 plus BP therapy and 43 participants in the control group receiving BP treatment only. Outcome Measures: The study measured preparation time before treatment, symptom-onset-to-door (SOTD), duration of shock, length of stay in the intensive care unit (ICU), and incidence of adverse reactions. The study also assessed changes between baseline and postintervention in inflammatory cytokines (ICs), immunological function, and myocardial-function markers. Finally, the research team conducted a one-year follow-up to determine participants' prognostic survival. Results: The groups showed no significant differences in the preparation time before treatment, SOTD, rescue success rate, and incidence of adverse events (P > .05), while the intervention group showed a significantly shorter duration of shock and length stay in the ICU and a significantly higher overall response rate (P < .05). The research team observed significant improvements in the T-lymphocyte subsets, ICs, and myocardial function in both groups postintervention, but the changes in the intervention group were significantly greater (P < .05). Follow-up results showed no significant differences in overall survival between the intervention and control groups (P > .05), but the average LC was significantly higher in the intervention group (P < .05). Conclusions: For SS patients, the combination of T-α1 and BP under the emergency GC can effectively improve their immunological and myocardial function, reduce inflammatory reaction, and prolong their LCs, which provides a greater guarantee of the effectiveness of treatment for SS patients in the future.


Subject(s)
Thymosin , Cytokines , Humans , Inflammation Mediators , Intensive Care Units , Thymalfasin/therapeutic use , Thymosin/therapeutic use
18.
Int Immunopharmacol ; 111: 109166, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35994852

ABSTRACT

INTRODUCTION: Heated intraperitoneal chemotherapy (HIPEC) is currently implemented in the treatment of peritoneal metastases from colorectal carcinoma (PM-CRC) origin. However, recurrence is common and the effectiveness of HIPEC has been questioned. The aim of this study was to evaluate the use of thymosin alpha 1 (Tα1), an immunomodulatory molecule, as an adjuvant to HIPEC treatment. METHODS: We developed an experimental model of HIPEC by the induction of PM-CRC in C57BL mice and intra-abdominal perfusion of mitomycin C (MMC). Mice were treated with Tα1 at 0.6 mg/kg for 5 days after HIPEC. Clinical and immunological parameters were compared between HIPEC and HIPEC + Tα1 groups. RESULTS: Treatment with Tα1 increased overall survival of mice compared to HIPEC treatment alone and sham-treated animals (16.1 ± 0.8 vs. 14.1 ± 0.6 and 11.8 ± 0.8, respectively, p = 0.02). Tα1 had no direct anti-tumor effect, as seen by lack of inhibition of tumor cell proliferation. Tα1 treatment induced a T helper (Th) 1 immune response in tumor metastases as evidenced by a significant increase of the Th1-specific markers IFN-γ and T-bet (1.21 ± 0.3 vs. 0.52 ± 0.08, p < 0.05; 0.88 ± 0.04 vs. 0.64 ± 0.14, p < 0.05, respectively). This Th1 skew was accompanied by increased CD8+ infiltration into omental and visceral metastases by Tα1 treatment compared to sham and HIPEC-treated animals (21.24 ± 2.16 vs. 10.45 ± 0.89 and 7.7 ± 1.3, p < 0.001; 14.12 ± 1.54 vs. 12.12 ± 0.01 and 6.64 ± 0.87, p < 0.01, respectively). CONCLUSIONS: Tα1 augments the effect of HIPEC by the induction of a Th1 anti-tumor immune response. Further experiments should evaluate Tα1 and other novel immunomodulators in order to exploit the immunological opportunities created by HIPEC.


Subject(s)
Carcinoma , Colonic Neoplasms , Colorectal Neoplasms , Hyperthermia, Induced , Peritoneal Neoplasms , Animals , Carcinoma/therapy , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Combined Modality Therapy , Hyperthermic Intraperitoneal Chemotherapy , Mice , Mice, Inbred C57BL , Models, Theoretical , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Thymalfasin/therapeutic use
19.
Cell Mol Biol (Noisy-le-grand) ; 67(6): 228-235, 2022 Feb 27.
Article in English | MEDLINE | ID: mdl-35818192

ABSTRACT

This study aimed to research the clinical effect of Xuebijing combined with thymosin α1 on patients with severe pneumonia complicated with sepsis, and its effect on serum inflammatory factors. For this purpose, 81 cases of severe pneumonia complicated with sepsis were collected. All patients were given early treatments. 41 cases who received Xuebijing injection by intravenous drip were selected as the control group. 40 cases who were treated through subcutaneous injection of thymosin α1 based on Xuebijing injection by intravenous drip were selected as the study group. The body temperature, respiration, heart rate, leukocytes, other general conditions, blood gas indexes, serum IL-6, TNF-α and CRP levels, bacterial clearance rate and therapy effect were recorded and compared before and after treatment. Results showed that after treatment, the body temperature, respiration, heart rate, leukocytes and other general conditions of the study group were lower than those in the control group (all p<0.05). The blood gas indexes pH and PaCO2 levels of the study group were lower than those of the control group. The levels of serum interleukin-6 (IL-6), serum tumor necrosis factor α (TNF-α) and C-reactive protein (CRP) in the study group were lower than those in the control group (all p<0.05). The bacterial clearance rate of the study group was lower than that of the control group (all p<0.05). The total effective rate of treatment of patients in the study group was higher than that of patients in the control group (all p<0.05). In general, Thymosin α1 and Xuebijing injection can improve the therapy effect of severe pneumonia complicated with sepsis, improve the hemorheology condition of patients, effectively remove bacteria and reduce the expression level of serum CRP, TNF-α, IL-6, IL-8 and other inflammatory factors in patients, which is worthy of clinical promotion.


Subject(s)
Drugs, Chinese Herbal , Pneumonia , Sepsis , Drugs, Chinese Herbal/therapeutic use , Humans , Inflammation/drug therapy , Interleukin-6 , Pneumonia/complications , Pneumonia/drug therapy , Sepsis/complications , Sepsis/drug therapy , Thymalfasin/therapeutic use , Tumor Necrosis Factor-alpha
20.
Int J Radiat Oncol Biol Phys ; 114(3): 433-443, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35870709

ABSTRACT

PURPOSE: To evaluate the efficacy of thymosin α1 in management of radiation pneumonitis (RP) in patients with locally advanced non-small cell lung cancer (LANSCLC) treated with concurrent chemoradiotherapy (CCRT). METHODS AND MATERIALS: This phase 2, single-arm trial enrolled patients with unresectable LANSCLC of 18 to 75 years' old and an Eastern Cooperative Oncology Group performance status of 0 to 1. Eligible patients received definitive CCRT and weekly thymosin α1 from the start of CCRT until 2 months after CCRT. Patients were administered 51 Gy in 17 daily fractions or 40 Gy in 10 daily fractions in the first course followed by a re-evaluation and those patients without disease progression had an adaptive plan of 15 Gy in 5 daily fractions or 24 Gy in 6 daily fractions as a boost. Concurrent chemotherapy consisted of weekly docetaxel (25 mg/m2) and nedaplatin (25 mg/m2) during radiation therapy. The primary endpoint was the incidence of Grade (G) ≥2 RP. Secondary endpoints included the incidence of late pulmonary fibrosis, total lymphocyte count (TLC), serum C-reactive protein (CRP) levels, and the composition of gut microbiota. TLC and CRP data were collected at baseline, 2 to 3 weeks during CCRT, the end of CCRT, 2 and 6 months after CCRT. Fecal samples were collected at baseline and the end of CCRT. Patients treated with CCRT but without thymosin α1 intervention during the same period were selected as the control group by the propensity score matching method. RESULTS: Sixty-nine patients were enrolled in the study, and another 69 patients were selected as the control group. The incidence of G≥2 RP was lower in the study group compared with control cases (36.2% vs 53.6%, P = .040). G1 late pulmonary fibrosis occurred in 2 (3.7%) patients of the control group compared with no event in the study group (P = .243). Compared with the control group, the incidence of G3 to G4 lymphopenia (19.1% vs 62.1%, P < .001) was lower, and the median TLC nadir (0.51 k/µL vs 0.30 k/µL, P < .001) was higher in the study group. The proportion of patients with maximum CRP ≥100 mg/L was lower in the study group (13.8% vs 29.7% P = .029). The diversity and community composition of the gut microbiota were not significantly different between the 2 groups. CONCLUSIONS: Administration of thymosin α1 during and after CCRT was associated with significant reductions in G≥2 RP and G3 to G4 lymphopenia in patients with LANSCLC compared with historic controls.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Lymphopenia , Pulmonary Fibrosis , Radiation Pneumonitis , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , C-Reactive Protein , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Docetaxel/therapeutic use , Humans , Lung Neoplasms/radiotherapy , Lymphopenia/etiology , Pulmonary Fibrosis/etiology , Radiation Pneumonitis/drug therapy , Radiation Pneumonitis/etiology , Thymalfasin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...