Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Immunol ; 393-394: 104782, 2023.
Article in English | MEDLINE | ID: mdl-37931572

ABSTRACT

Type I interferons (IFN), especially human IFN alpha (IFNα), have been utilized for antitumor therapy for decades. Human interferon beta (IFNß) is rarely used for cancer treatment, despite advantages over IFNα in biological activities such as tumor growth inhibition and dendritic cell (DC) activation. The utilization of pegylated human IFNß (PEG-IFNß), as monotherapy or in combination with immune checkpoint inhibitors (ICIs) was evaluated in this study through in vivo efficacy studies in syngeneic mouse melanoma, non-small cell lung cancer (NSCLC), and colon adenocarcinoma (COAD) models resistant to immune checkpoint inhibitors (ICIs). In vitro comparative study of PEG-IFNß and pegylated IFNα-2b was performed in terms of tumor growth inhibition against human melanoma, NSCLC and COAD cell lines and activation of human monocyte-derived DCs (MoDCs). Our data demonstrate that the in vivo antitumor effects of PEG-IFNß are partially attributable to tumor growth-inhibitory effects and DC-activating activities, superior to pegylated IFNα-2b. Our findings suggest that utilizing PEG-IFNß as an antitumor therapy can enhance the therapeutic effect of ICIs in ICI-resistant tumors by directly inhibiting tumor growth and induction of DC maturation.


Subject(s)
Adenocarcinoma , Carcinoma, Non-Small-Cell Lung , Colonic Neoplasms , Lung Neoplasms , Melanoma , Animals , Mice , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Adenocarcinoma/drug therapy , Lung Neoplasms/pathology , Colonic Neoplasms/drug therapy , Interferon-alpha/pharmacology , Interferon alpha-2/therapeutic use , Melanoma/drug therapy , Interferon-beta/metabolism , Interferon-beta/therapeutic use , Polyethylene Glycols/therapeutic use , Dendritic Cells/metabolism
2.
Front Immunol ; 14: 1143796, 2023.
Article in English | MEDLINE | ID: mdl-37033979

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV), an emerging bunyavirus, causes severe fever with thrombocytopenia syndrome (SFTS), with a high fatality rate of 20%-30%. At present, however, the pathogenesis of SFTSV remains largely unclear and no specific therapeutics or vaccines against its infection are currently available. Therefore, animal models that can faithfully recapitulate human disease are important to help understand and treat SFTSV infection. Here, we infected seven Chinese rhesus macaques (Macaca mulatta) with SFTSV. Virological and immunological changes were monitored over 28 days post-infection. Results showed that mild symptoms appeared in the macaques, including slight fever, thrombocytopenia, leukocytopenia, increased aspartate aminotransferase (AST) and creatine kinase (CK) in the blood. Viral replication was persistently detectable in lymphoid tissues and bone marrow even after viremia disappeared. Immunocyte detection showed that the number of T cells (mainly CD8+ T cells), B cells, natural killer (NK) cells, and monocytes decreased during infection. In detail, effector memory CD8+ T cells declined but showed increased activation, while both the number and activation of effector memory CD4+ T cells increased significantly. Furthermore, activated memory B cells decreased, while CD80+/CD86+ B cells and resting memory B cells (CD27+CD21+) increased significantly. Intermediate monocytes (CD14+CD16+) increased, while myeloid dendritic cells (mDCs) rather than plasmacytoid dendritic cells (pDCs) markedly declined during early infection. Cytokines, including interleukin-6 (IL-6), interferon-inducible protein-10 (IP-10), and macrophage inflammatory protein 1 (MCP-1), were substantially elevated in blood and were correlated with activated CD4+ T cells, B cells, CD16+CD56+ NK cells, CD14+CD16+ monocytes during infection. Thus, this study demonstrates that Chinese rhesus macaques infected with SFTSV resemble mild clinical symptoms of human SFTS and provides detailed virological and immunological parameters in macaques for understanding the pathogenesis of SFTSV infection.


Subject(s)
Phlebovirus , Severe Fever with Thrombocytopenia Syndrome , Animals , Humans , Macaca mulatta , CD8-Positive T-Lymphocytes , Cytokines
3.
Sci China Life Sci ; 66(8): 1818-1830, 2023 08.
Article in English | MEDLINE | ID: mdl-36598621

ABSTRACT

Multivalent vaccines combining crucial mutations from phylogenetically divergent variants could be an effective approach to defend against existing and future SARS-CoV-2 variants. In this study, we developed a tetravalent COVID-19 vaccine SCTV01E, based on the trimeric Spike protein of SARS-CoV-2 variants Alpha, Beta, Delta, and Omicron BA.1, with a squalene-based oil-in-water adjuvant SCT-VA02B. In the immunogenicity studies in naïve BALB/c and C57BL/6J mice, SCTV01E exhibited the most favorable immunogenic characteristics to induce balanced and broad-spectrum neutralizing potencies against pre-Omicron variants (D614G, Alpha, Beta, and Delta) and newly emerging Omicron subvariants (BA.1, BA.1.1, BA.2, BA.3, and BA.4/5). Booster studies in C57BL/6J mice previously immunized with D614G monovalent vaccine demonstrated superior neutralizing capacities of SCTV01E against Omicron subvariants, compared with the D614G booster regimen. Furthermore, SCTV01E vaccination elicited naïve and central memory T cell responses to SARS-CoV-2 ancestral strain and Omicron spike peptides. Together, our comprehensive immunogenicity evaluation results indicate that SCTV01E could become an important COVID-19 vaccine platform to combat surging infections caused by the highly immune evasive BA.4/5 variants. SCTV01E is currently being studied in a head-to-head immunogenicity comparison phase 3 clinical study with inactivated and mRNA vaccines (NCT05323461).


Subject(s)
COVID-19 , SARS-CoV-2 , Mice , Animals , Humans , Mice, Inbred C57BL , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19 Vaccines , Vaccines, Combined , Squalene , Antibodies, Neutralizing , Antibodies, Viral
4.
Vaccine ; 39(6): 915-925, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33451779

ABSTRACT

Zika virus (ZIKV) infection has caused major public health problems recently. To develop subunit vaccines for ZIKV, we have previously constructed recombinant ZIKV envelope protein domain III (EDIII), and the entire ectodomain (E80, which comprises EDI, EDII and EDIII), as vaccine candidates and showed both of them being immunogenic and protective in murine models. In this follow-up study, we compared these vaccine candidates in non-human primates. Both of them elicited neutralizing antibody responses, but only E80 immunization inhibited ZIKV infection in both peripheral blood and monkey tissues, whereas EDIII increased blood ZIKV RNA through possibly antibody-dependent enhancement. Further investigations revealed that the virion-binding antibody response in E80 immunized monkeys persisted longer and stronger than in EDIII immunized monkeys. These results demonstrate that E80 is superior to EDIII as a vaccine candidate, and that the magnitude, quality and durability of virion-binding neutralizing antibodies are correlates of protection.


Subject(s)
Viral Vaccines/immunology , Zika Virus Infection , Animals , Antibodies, Neutralizing , Antibodies, Viral , Antibody Formation , Follow-Up Studies , Macaca mulatta , Recombinant Proteins/immunology , Viral Envelope Proteins/genetics , Zika Virus , Zika Virus Infection/prevention & control
5.
J Vet Pharmacol Ther ; 42(6): 660-664, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31222770

ABSTRACT

Altrenogest, a synthetic progestogen, is characterized by its estrus synchronization in mares, ewes, sows, and gilts. To investigate the pharmacokinetic profile and evaluate its accumulation in gilts, 18 oral doses of 20 mg altrenogest/gilt/day were given to eight healthy gilts at an interval of 24 hr. Plasma samples were collected, and altrenogest was determined by ultra-high-performance liquid chromatography with mass spectrometry. WinNonlin 6.4 software was used to calculate the pharmacokinetic parameters through noncompartmental model analysis. After the first administration (D 1), the pharmacokinetic parameters, including Tmax , Cmax , and the elimination half-life (T1/2λz ), were similar to those observed after the final administration (D 18). However, the mean residence time at D 1 was significantly lower than D 18. As a whole, the mean steady-state plasma concentration (Css ), degree fluctuation (DF), accumulation factor (Rac ), and area under the plasma concentration-time curve in steady state (AUCss ) were 22.69 ± 6.15 ng/ml, 270.64 ± 42.51%, 1.53 ± 0.23, and 544.63 ± 147.49 ng hr/ml, respectively. These results showed that after 18 consecutive days of oral administration of altrenogest, plasma concentrations of altrenogest had a certain degree of fluctuation, without significant accumulations.


Subject(s)
Progesterone Congeners/pharmacokinetics , Swine/blood , Trenbolone Acetate/analogs & derivatives , Administration, Oral , Animals , Area Under Curve , Female , Half-Life , Progesterone Congeners/blood , Trenbolone Acetate/administration & dosage , Trenbolone Acetate/blood , Trenbolone Acetate/pharmacokinetics
6.
J Vet Pharmacol Ther ; 42(1): 111-115, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30393987

ABSTRACT

The pharmacokinetic parameters of moxidectin (MXD) after intravenous and pour-on (topical) administration were studied in sixteen pigs at a single dose of 1.25 and 2.5 mg/kg BW (body weight), respectively. Blood samples were collected at pretreatment time (0 hr) over 40 days. The plasma kinetics were analyzed by WinNonlin 6.3 software through a noncompartmental model. For intravenous administration (n = 8), the elimination half-life (λZ ), the apparent volume of distribution (Vz ), and clearance (Cl) were 10.29 ± 1.90 days, 89.575 ± 29.856 L/kg, and 5.699 ± 2.374 L/kg, respectively. For pour-on administration (n = 8), the maximum plasma drug concentration (Cmax ), time to maximum plasma concentration (Tmax ), and λZ were 7.49 ng/ml, 1.72, and 6.20 days, respectively. MXD had a considerably low absolute pour-on bioavailability of 9.2%, but the mean residence time (MRT) for pour-on administration 10.88 ± 1.75 days was longer than 8.99 ± 2.48 days for intravenous administration. These results showed that MXD was absorbed via skin rapidly and eliminated slowly. The obtained data might contribute to refine the dosage regime for topical MXD administration.


Subject(s)
Antiparasitic Agents/pharmacokinetics , Macrolides/pharmacokinetics , Swine/metabolism , Administration, Cutaneous , Animals , Antiparasitic Agents/administration & dosage , Antiparasitic Agents/blood , Half-Life , Injections, Intravenous/veterinary , Macrolides/administration & dosage , Macrolides/blood , Male , Swine/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...