58 research outputs found

    Latent Membrane Protein 1 as a molecular adjuvant for single-cycle lentiviral vaccines

    Get PDF
    Background Molecular adjuvants are a promising method to enhance virus-specific immune responses and protect against HIV-1 infection. Immune activation by ligands for receptors such as CD40 can induce dendritic cell activation and maturation. Here we explore the incorporation of two CD40 mimics, Epstein Barr Virus gene LMP1 or an LMP1-CD40 chimera, into a strain of SIV that was engineered to be limited to a single cycle of infection. Results Full length LMP1 or the chimeric protein LMP1-CD40 was cloned into the nef-locus of single-cycle SIV. Human and Macaque monocyte derived macrophages and DC were infected with these viruses. Infected cells were analyzed for activation surface markers by flow cytometry. Cells were also analyzed for secretion of pro-inflammatory cytokines IL-1ÎČ, IL-6, IL-8, IL-12p70 and TNF by cytometric bead array. Conclusions Overall, single-cycle SIV expressing LMP1 and LMP1-CD40 produced a broad and potent TH_H1-biased immune response in human as well as rhesus macaque macrophages and DC when compared with control virus. Single-cycle SIV-LMP1 also enhanced antigen presentation by lentiviral vector vaccines, suggesting that LMP1-mediated immune activation may enhance lentiviral vector vaccines against HIV-1

    Experiment for cryogenic large-aperture intensity mapping: instrument design

    Get PDF
    The experiment for cryogenic large-aperture intensity mapping (EXCLAIM) is a balloon-borne telescope designed to survey star formation in windows from the present to z  =  3.5. During this time, the rate of star formation dropped dramatically, while dark matter continued to cluster. EXCLAIM maps the redshifted emission of singly ionized carbon lines and carbon monoxide using intensity mapping, which permits a blind and complete survey of emitting gas through statistics of cumulative brightness fluctuations. EXCLAIM achieves high sensitivity using a cryogenic telescope coupled to six integrated spectrometers employing kinetic inductance detectors covering 420 to 540 GHz with spectral resolving power R  =  512 and angular resolution ≈4  arc min. The spectral resolving power and cryogenic telescope allow the survey to access dark windows in the spectrum of emission from the upper atmosphere. EXCLAIM will survey 305  deg2 in the Sloan Digital Sky Survey Stripe 82 field from a conventional balloon flight in 2023. EXCLAIM will also map several galactic fields to study carbon monoxide and neutral carbon emission as tracers of molecular gas. We summarize the design phase of the mission

    31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016) : part two

    Get PDF
    Background The immunological escape of tumors represents one of the main ob- stacles to the treatment of malignancies. The blockade of PD-1 or CTLA-4 receptors represented a milestone in the history of immunotherapy. However, immune checkpoint inhibitors seem to be effective in specific cohorts of patients. It has been proposed that their efficacy relies on the presence of an immunological response. Thus, we hypothesized that disruption of the PD-L1/PD-1 axis would synergize with our oncolytic vaccine platform PeptiCRAd. Methods We used murine B16OVA in vivo tumor models and flow cytometry analysis to investigate the immunological background. Results First, we found that high-burden B16OVA tumors were refractory to combination immunotherapy. However, with a more aggressive schedule, tumors with a lower burden were more susceptible to the combination of PeptiCRAd and PD-L1 blockade. The therapy signifi- cantly increased the median survival of mice (Fig. 7). Interestingly, the reduced growth of contralaterally injected B16F10 cells sug- gested the presence of a long lasting immunological memory also against non-targeted antigens. Concerning the functional state of tumor infiltrating lymphocytes (TILs), we found that all the immune therapies would enhance the percentage of activated (PD-1pos TIM- 3neg) T lymphocytes and reduce the amount of exhausted (PD-1pos TIM-3pos) cells compared to placebo. As expected, we found that PeptiCRAd monotherapy could increase the number of antigen spe- cific CD8+ T cells compared to other treatments. However, only the combination with PD-L1 blockade could significantly increase the ra- tio between activated and exhausted pentamer positive cells (p= 0.0058), suggesting that by disrupting the PD-1/PD-L1 axis we could decrease the amount of dysfunctional antigen specific T cells. We ob- served that the anatomical location deeply influenced the state of CD4+ and CD8+ T lymphocytes. In fact, TIM-3 expression was in- creased by 2 fold on TILs compared to splenic and lymphoid T cells. In the CD8+ compartment, the expression of PD-1 on the surface seemed to be restricted to the tumor micro-environment, while CD4 + T cells had a high expression of PD-1 also in lymphoid organs. Interestingly, we found that the levels of PD-1 were significantly higher on CD8+ T cells than on CD4+ T cells into the tumor micro- environment (p < 0.0001). Conclusions In conclusion, we demonstrated that the efficacy of immune check- point inhibitors might be strongly enhanced by their combination with cancer vaccines. PeptiCRAd was able to increase the number of antigen-specific T cells and PD-L1 blockade prevented their exhaus- tion, resulting in long-lasting immunological memory and increased median survival

    Constitutively Active MAVS Inhibits HIV-1 Replication via Type I Interferon Secretion and Induction of HIV-1 Restriction Factors.

    No full text
    Type I interferon is known to inhibit HIV-1 replication through the induction of interferon stimulated genes (ISG), including a number of HIV-1 restriction factors. To better understand interferon-mediated HIV-1 restriction, we constructed a constitutively active form of the RIG-I adapter protein MAVS. Constitutive MAVS was generated by fusion of full length MAVS to a truncated form of the Epstein Barr virus protein LMP1 (ΔLMP1). Supernatant from ΔLMP1-MAVS-transfected 293T cells contained high levels of type I interferons and inhibited HIV replication in both TZM-bl and primary human CD4+ T cells. Supernatant from ΔLMP1-MAVS-transfected 293T cells also inhibited replication of VSV-G pseudotyped single cycle SIV in TZM-bl cells, suggesting restriction was post-entry and common to both HIV and SIV. Gene array analysis of ΔLMP1-MAVS-transfected 293T cells and trans-activated CD4+ T cells showed significant upregulation of ISG, including previously characterized HIV restriction factors Viperin, Tetherin, MxB, and ISG56. Interferon blockade studies implicated interferon-beta in this response. In addition to direct viral inhibition, ΔLMP1-MAVS markedly enhanced secretion of IFN-ÎČ and IL-12p70 by dendritic cells and the activation and maturation of dendritic cells. Based on this immunostimulatory activity, an adenoviral vector (Ad5) expressing ΔLMP1-MAVS was tested as a molecular adjuvant in an HIV vaccine mouse model. Ad5-Gag antigen combined with Ad5-ΔLMP1-MAVS enhanced control of vaccinia-gag replication in a mouse challenge model, with 4/5 animals showing undetectable virus following challenge. Overall, ΔLMP1-MAVS is a promising reagent to inhibit HIV-1 replication in infected tissues and enhance vaccine-mediated immune responses, while avoiding toxicity associated with systemic type I interferon administration

    Abstract B062: Immune activation by LMP1 CD40 pathway and LMP1-IPS-1 STING pathway activators

    No full text
    Abstract The strongest CD8+ T cell responses ever recorded in humans occur as a result of infection by the Epstein-Barr Virus (EBV). Following a massive CD8+ T cell response during acute EBV infection, ∌5.5% of the total CD8+ T cell repertoire in blood is thereafter committed to the life-long suppression of viral replication. The key to this response is the EBV Latent Membrane Protein-1 (LMP1) which functions as a constitutively activated form of the CD40 receptor. LMP1 consists of an N-terminal domain that clusters in cell membranes and a C-terminal domain that engages the proteins of the CD40 signalosome. Studies to determine if LMP1 could be used for immunotherapy led to the following conclusions: (1) LMP1 mRNA directly activated human dendritic cells (DCs), showing its potential as an adjuvant for mRNA vaccines; (2) Adenovirus (Ad)-delivered LMP1 strongly enhanced vaccine-induced CD8+ T cell responses in mice, indicating that LMP1 may enhance Ad-based oncolytic therapies; (3) lentiviruses incorporating LMP1 became strong activators of DCs in vitro; and (4) DNA vaccines incorporating LMP1 protected mice from tumor challenge in a B16F10 melanoma metastasis model. These data show that the LMP1 nucleic acid sequence is a portable genetic adjuvant with broad applicability to tumor immunotherapy. In related studies, the membrane clustering N-terminal domain of LMP1 was fused to Interferon-beta Promoting Stimulator-1 (IPS-1, also called MAVS, VISA, or Cardif), a protein that requires membrane clustering to become activated. This fusion protein acts independently of cyclic dinucleotides (CDN) to activate downstream mediators of the STING pathway. Vaccination of mice with Ad expressing antigen plus LMP1-IPS-1 resulted in complete protection from challenge with Vaccinia-antigen virus (&amp;gt;1 million-fold protection). These data suggest that LMP1-IPS-1 combined with gene delivery technologies may supplant the need for small molecule CDNs for STING pathway activation in immuno-oncology. Citation Format: Richard S. Kornbluth, Sachin Gupta, James M. Termini, Elizabeth Guirado, Geoffrey W. Stone. Immune activation by LMP1 CD40 pathway and LMP1-IPS-1 STING pathway activators [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr B062.</jats:p

    A multi-trimeric fusion of CD40L and gp100 tumor antigen activates dendritic cells and enhances survival in a B16-F10 melanoma DNA vaccine model

    No full text
    ‱DNA vaccine technology fusing cancer antigens to CD40L soluble multi-trimers.‱Allows targeting of vaccine antigens to dendritic cells in situ.‱Activates and matures dendritic cells.‱Vaccine induces an anti-tumor immune response when combined with IL-12 and GM-CSF. Vaccination with tumor-associated antigens can induce cancer-specific CD8+ T cells. A recent improvement has been the targeting of antigen to dendritic cells (DC) using antibodies that bind DC surface molecules. This study explored the use of multi-trimers of CD40L to target the gp100 melanoma tumor antigen to DC. The spontaneously-multimerizing gene Surfactant Protein D (SPD) was used to fuse gp100 tumor antigen and CD40L, creating the recombinant protein SPD-gp100-CD40L. This “third generation” DC-targeting vaccine was designed to both target antigen to DC and optimally activate dendritic cells by aggregating CD40 trimers on the DC membrane surface. SPD-gp100-CD40L expressed as a 110kDa protein. Analytical light scattering analysis gave elution data corresponding to 4-trimer and multi-trimer SPD-gp100-CD40L oligomers. The protein was biologically active on dendritic cells and induced CD40-mediated NF-ÎșB signaling. DNA vaccination with SPD-gp100-CD40L plasmid, together with plasmids encoding IL-12p70 and GM-CSF, significantly enhanced survival and inhibited tumor growth in a B16-F10 melanoma model. Expression of gp100 and SPD-CD40L as separate molecules did not enhance survival, highlighting the requirement to encode gp100 within SPD-CD40L for optimal vaccine activity. These data support a model where DNA vaccination with SPD-gp100-CD40L targets gp100 to DC in situ, induces activation of these DC, and generates a protective anti-tumor response when given in combination with IL-12p70 and GM-CSF plasmids

    Beta-interferon mediates inhibition of HIV-1 BaL replication.

    No full text
    <p>(A) The relative level of HIV-1 BaL strain viral replication in TZM-bl cells was measured following incubation with supernatant from 293T cells transfected with either pcDNA3.1 or pΔLMP1-MAVS plasmid, combined with 60 ÎŒg of isotype control antibody, anti-IFN-ÎČ antibody, or anti-IFN-α antibody. (B) Human CD4+ T cells were infected with HIV-1 BaL in the presence of increasing concentrations of interferon-α and compared to infection in the presence of 293T supernatant following transfection with either pcDNA3.1 or pΔLMP1-MAVS plasmid. (C) Supernatant from 293T cells transfected with pcDNA3.1 GFP, LMP1, or ΔLMP1-MAVS plasmid was assayed for IFN-α and IFN-ÎČ secretion by ELISA. NT: no treatment.</p
    • 

    corecore