Product Citations: 13

Development of an Intranasally- and Intramuscularly-Administrable Replicon Vaccine Efficacious Against H5N1 Influenza Virus

Preprint on BioRxiv : the Preprint Server for Biology on 5 April 2025 by McClary, W. D., Brandt, D. S., et al.

ABSTRACT The risk of a respiratory viral pandemic is significant, including from the now widespread panzootic H5N1 influenza virus, highlighting the need for effective, stable, and inexpensive vaccine technologies that elicit strongly protective immunity. Intranasal vaccines can stimulate local immune responses at the site of natural respiratory viral infection, a key characteristic that can not only reduce morbidity and mortality caused by respiratory viruses but also potentially reduce viral transmissibility to limit outbreaks. Nucleic acid vaccines are now a valuable tool in pandemic responses, with high potency and rapid adaptability to target circulating or emerging viral strains; however, data are limited on which vaccine attributes are needed for efficient transmucosal delivery and immune stimulation following intranasal delivery. To demonstrate proof of concept, here we have developed a replicon vaccine expressing an H5 influenza antigen that uses a nanostructured lipid carrier (NLC) delivery system. A relationship was established between the molar ratio of positive charges on the NLC to the negative charges on the nucleic acid (N:P ratio) and the immunogenicity of the vaccine formulations, with higher N:P ratios resulting in an increase in vaccine immunogenicity. We demonstrated the ability of this replicon vaccine to be administered via intramuscular and intranasal routes with a singular vaccine formulation. The vaccine induced systemic immunity when dosed intramuscularly or intranasally in an immunocompetent mouse model, whereas intranasal dosing uniquely stimulated a strong mucosal immune response. Moreover, a mixed intramuscular/intranasal dosing strategy using this unified formulation stimulated a balanced systemic and mucosal immune response. Finally, we demonstrated the protective efficacy of this intranasally and intramuscularly/intranasally delivered H5 replicon-NLC vaccine against morbidity and mortality in a lethal H5N1 influenza challenge ferret model. This work establishes the replicon-NLC vaccine platform as a potential novel intranasal technology for rapid pandemic response.

  • Immunology and Microbiology

This research utilized network pharmacology to investigate the potential of Fuzheng Qudu prescription (FZQDP) in treating lung cancer (LC).
The components and their targets of FZQDP were analyzed for their relationship with LC-related targets using bioinformatics tools. Mouse Lewis lung carcinoma (LLC) cells were cultured in vitro and treated with FZQDP or cisplatin (DDP) before applying the MTT assay to determine FZQDP concentrations, and the IC50 value. According to the IC50 value, the effect of FZQDP on apoptosis and cell cycle was detected by flow cytometry. Mouse tumor growth was recorded using live animal imaging, and measurements of tumor and spleen weight were used to calculate the tumor inhibition rate and spleen index. The effects on mouse liver and kidneys were observed by analyzing levels of AST, ALT, BUN, and CRE in blood and hematoxylin and eosin (H & E) stained sections. Additionally, levels of IL-2, IL-10, IL-6, and IFN-γ in serum, along with the frequencies of CD4+ and CD8+ T cells in the spleen, were measured using Mouse multiple Cytokine Assay and flow cytometry, respectively.
SRC, STAT3, MAPK3, and MAPK1 could be crucial targets of FZQDP in the treatment of LC. FZQDP demonstrated inhibition of LC cell proliferation and tumor growth, as well as enhancement of apoptosis and induction of G2 phase cell cycle arrest. Furthermore, FZQDP led to elevated levels of IL-2 and IFN-γ, increased frequencies of CD4+ T cells and decreased levels of IL-6 and IL-10. Importantly, FZQDP did not exhibit any noticeable hepatotoxic or nephrotoxic effects in mice.
FZQDP may target multiple signaling pathways to treat LC. In a LC mouse model, FZQDP was found to inhibit tumor growth and improve immune function.
© 2024 The Authors.

  • Cancer Research
  • Pharmacology

One-carbon unit supplementation fuels tumor-infiltrating T cells and augments checkpoint blockade

Preprint on BioRxiv : the Preprint Server for Biology on 3 November 2023 by Xu, X., Chen, Z., et al.

Summary Nucleotides perform important metabolic functions, carrying energy and feeding nucleic acid synthesis. Here, we use isotope tracing-mass spectrometry to quantitate the contributions to purine nucleotides of salvage versus de novo synthesis. We further explore the impact of augmenting a key precursor for purine synthesis, one-carbon (1C) units. We show that tumors and tumor-infiltrating T cells (relative to splenic T cells) synthesize purines de novo . Purine synthesis requires two 1C units, which come from serine catabolism and circulating formate. Shortage of 1C units is a potential bottleneck for anti-tumor immunity. Elevating circulating formate drives its usage by tumor-infiltrating T cells. Orally administered methanol functions as a formate pro-drug, with deuteration enabling control of formate-production kinetics. In MC38 tumors, safe doses of methanol raise formate levels and augment anti-PD-1 checkpoint blockade, tripling durable regressions. Thus, 1C deficiency can gate antitumor immunity and this metabolic checkpoint can be overcome with pharmacological 1C supplementation. Statement of significance Checkpoint blockade has revolutionized cancer therapy. Durable tumor control, however, is achieved in only a minority of patients. We show that the efficacy of anti-PD-1 blockade can be enhanced by metabolic supplementation with one-carbon donors. Such donors support nucleotide synthesis in tumor-infiltrating T cells and merit future clinical evaluation.

  • Cancer Research
  • Immunology and Microbiology

Co-engagement of TIGIT+immune cells to PD-L1+tumours by a bispecific antibody potentiates T-cell response and tumour control

Preprint on BioRxiv : the Preprint Server for Biology on 14 May 2023 by Cui, X., Zhu, X., et al.

Co-targeting PD-1/PD-L1 and TIGIT/CD226 is being pursued to broaden the efficacy of current immunotherapy. Here we demonstrate that a bispecific antibody (BsAb) targeting PD-L1 and TIGIT, HB0036, shows major advantages over the combination of the two parental monoclonal Abs (mAbs). We demonstrated that HB0036 co-engages PD-L1 + tumour cells and TIGIT + T cells, and thereby upregulates CD226 on T cells and induces a greater T-cell proliferative response compared to the combination of the parental antibodies in vitro . In vivo , HB0036 recruits greater amounts of TIGIT antibody in PD-L1 + tumours but not in PD-L1 - tumours, compared with therapy using the two parental antibodies. We also observed improved tumour control and favourable immunological signatures with HB0036 in syngeneic and xenograft tumour models. Collectively, these findings demonstrate that bispecific antibodies targeting PD-L1 and TIGIT offer superior benefits in cancer immunotherapy compared with therapy using the two parental antibodies. Based on these studies, a phase I clinical trial with HB0036 has been initiated in patients with solid tumours ( NCT05417321 ).

  • Cancer Research
  • Immunology and Microbiology

Oral IRAK4 inhibitor BAY-1834845 prevents acute respiratory distress syndrome.

In Biomedicine Pharmacotherapy = Biomédecine Pharmacothérapie on 1 September 2022 by Li, Q., Li, R., et al.

Acute respiratory distress syndrome (ARDS) is a lethal clinical entity that has become an emergency event with the outbreak of COVID-19. However, to date, there are no well-proven pharmacotherapies except dexamethasone. This study is aimed to evaluate IRAK4 inhibitors as a potential treatment for ARDS-cytokine release syndrome (CRS). We applied two IRAK4 inhibitors, BAY-1834845 and PF-06650833 to an inhaled lipopolysaccharide (LPS)-induced ARDS mouse model with control of high dose dexamethasone (10 mg/kg). Unexpectedly, although both compounds had excellent IC50 on IRAK4 kinase activity, only BAY-1834845 but not PF-06650833 or high dose dexamethasone could significantly prevent lung injury according to a blinded pathology scoring. Further, only BAY-1834845 and BAY-1834845 combined with dexamethasone could effectively improve the injury score of pre-existed ARDS. Compared with PF-06650833 and high dose dexamethasone, BAY-1834845 remarkably decreased inflammatory cells infiltrating lung tissue and neutrophil count in BALF. BAY-1834845, DEX, and the combination of the two agents could decrease BALF total T cells, monocyte, and macrophages. In further cell type enrichment analysis based on lung tissue RNA-seq, both BAY-1834845 and dexamethasone decreased signatures of inflammatory cells and effector lymphocytes. Interestingly, unlike the dexamethasone group, BAY-1834845 largely preserved the signatures of naïve lymphocytes and stromal cells such as endothelial cells, chondrocytes, and smooth muscle cells. Differential gene enrichment suggested that BAY-1834845 downregulated genes more efficiently than dexamethasone, especially TNF, IL-17, interferon, and Toll-like receptor signaling.
Copyright © 2022. Published by Elsevier Masson SAS.

  • FC/FACS
  • Mus musculus (House mouse)
View this product on CiteAb