CD40 ligand/interferon-γ matured DC immunization with gp100 antigen HLA class I  A *0201 restricted peptides in patients with newly diagnosed metastatic melanoma.

2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 2525-2525
Author(s):  
Gerald P. Linette ◽  
Michelle Becker-Hapak ◽  
Alexander Huang ◽  
Amer Alyasiry ◽  
Megan Chan ◽  
...  

2525 Background: CD40L/IFN-γ matured Dendritic Cells (DCs) produce IL-12 and are potent antigen-presenting cells for naïve resting T cells. We sought to determine the magnitude and kinetics of CD8+ T cell growth in patients receiving autologous CD40L/IFN-γ matured DC and identify biomarkers associated with clinical outcome. Methods: A phase I clinical trial (NCT00683670) incorporating CD40L/IFN-γ for the ex vivo maturation of autologous DCs pulsed with three well characterized gp100 melanoma antigen derived peptides (G154, G209-2M, G280-9V) was initiated with enrollment from 2008-11 at a single center. HLA-A*0201+ individuals with treatment naïve metastatic melanoma were immunized every 3 weeks by intravenous infusion for six doses after a single dose of cyclophosphamide (300 mg/m2 iv). CT imaging was performed at baseline, week 9 and 18 for clinical assessment using RECIST. Responding patients were eligible for maintenance doses every 2-4 months. PBMC were taken weekly for immune monitoring by tetramer analysis and functional assays. DC preparations were characterized to assess for biomarkers of response. Results: 10 patients were screened. Among the 7 treated patients, there were 3 confirmed responses (independently verified), including one durable CR >3 years and 2 PR. Three patients had rapid disease progression and received only 3 doses. Four patients (1 CR, 2 PR, 1 PD) received 6 or more vaccine doses. No SAEs were noted. There was no correlation between tumor volume and response. Using pre-specified immune response criteria, 6 (86%) treated patients developed CD8+ T cell immunity to all three peptides as assessed by tetramer analysis. The vaccine-induced T cells from all 6 individuals were polyfunctional and killed gp100+, HLA-A2+ human melanoma targets in a standard 51Cr release assay. IL-12 production by DCs correlated with TTP (p=0.0198, likelihood ratio test) but not OS (p=0.08). Conclusions: Weekly immune monitoring reveals the rapid onset of CD8+ T cell immunity against gp100 among the responder patients. This is the first DC vaccine clinical trial in melanoma to demonstrate a correlation of IL-12 production and TTP.

2003 ◽  
Vol 198 (2) ◽  
pp. 267-279 ◽  
Author(s):  
Shin-ichiro Fujii ◽  
Kanako Shimizu ◽  
Caroline Smith ◽  
Laura Bonifaz ◽  
Ralph M. Steinman

The maturation of dendritic cells (DCs) allows these antigen-presenting cells to initiate immunity. We pursued this concept in situ by studying the adjuvant action of α-galactosylceramide (αGalCer) in mice. A single i.v. injection of glycolipid induced the full maturation of splenic DCs, beginning within 4 h. Maturation was manifest by marked increases in costimulator and major histocompatibility complex class II expression, interferon (IFN)-γ production, and stimulation of the mixed leukocyte reaction. These changes were not induced directly by αGalCer but required natural killer T (NKT) cells acting independently of the MyD88 adaptor protein. To establish that DC maturation was responsible for the adjuvant role of αGalCer, mice were given αGalCer together with soluble or cell-associated ovalbumin antigen. Th1 type CD4+ and CD8+ T cell responses developed, and the mice became resistant to challenge with ovalbumin-expressing tumor. DCs from mice given ovalbumin plus adjuvant, but not the non-DCs, stimulated ovalbumin-specific proliferative responses and importantly, induced antigen-specific, IFN-γ producing, CD4+ and CD8+ T cells upon transfer into naive animals. In the latter instance, immune priming did not require further exposure to ovalbumin, αGalCer, NKT, or NK cells. Therefore a single dose of αGalCer i.v. rapidly stimulates the full maturation of DCs in situ, and this accounts for the induction of combined Th1 CD4+ and CD8+ T cell immunity to a coadministered protein.


2017 ◽  
Vol 7 (1) ◽  
Author(s):  
Liangquan Zhu ◽  
Xinxin Zhao ◽  
Qing Yin ◽  
Xianyong Liu ◽  
Xiang Chen ◽  
...  

2018 ◽  
Vol 9 ◽  
Author(s):  
Cintia L. Araujo Furlan ◽  
Jimena Tosello Boari ◽  
Constanza Rodriguez ◽  
Fernando P. Canale ◽  
Facundo Fiocca Vernengo ◽  
...  

Vaccines ◽  
2020 ◽  
Vol 8 (4) ◽  
pp. 706
Author(s):  
Chunmei Fu ◽  
Li Zhou ◽  
Qing-Sheng Mi ◽  
Aimin Jiang

As the sentinels of the immune system, dendritic cells (DCs) play a critical role in initiating and regulating antigen-specific immune responses. Cross-priming, a process that DCs activate CD8 T cells by cross-presenting exogenous antigens onto their MHCI (Major Histocompatibility Complex class I), plays a critical role in mediating CD8 T cell immunity as well as tolerance. Current DC vaccines have remained largely unsuccessful despite their ability to potentiate both effector and memory CD8 T cell responses. There are two major hurdles for the success of DC-based vaccines: tumor-mediated immunosuppression and the functional limitation of the commonly used monocyte-derived dendritic cells (MoDCs). Due to their resistance to tumor-mediated suppression as inert vesicles, DC-derived exosomes (DCexos) have garnered much interest as cell-free therapeutic agents. However, current DCexo clinical trials have shown limited clinical benefits and failed to generate antigen-specific T cell responses. Another exciting development is the use of naturally circulating DCs instead of in vitro cultured DCs, as clinical trials with both human blood cDC2s (type 2 conventional DCs) and plasmacytoid DCs (pDCs) have shown promising results. pDC vaccines were particularly encouraging, especially in light of promising data from a recent clinical trial using a human pDC cell line, despite pDCs being considered tolerogenic and playing a suppressive role in tumors. However, how pDCs generate anti-tumor CD8 T cell immunity remains poorly understood, thus hindering their clinical advance. Using a pDC-targeted vaccine model, we have recently reported that while pDC-targeted vaccines led to strong cross-priming and durable CD8 T cell immunity, cross-presenting pDCs required cDCs to achieve cross-priming in vivo by transferring antigens to cDCs. Antigen transfer from pDCs to bystander cDCs was mediated by pDC-derived exosomes (pDCexos), which similarly required cDCs for cross-priming of antigen-specific CD8 T cells. pDCexos thus represent a new addition in our arsenal of DC-based cancer vaccines that would potentially combine the advantage of pDCs and DCexos.


2003 ◽  
Vol 198 (1) ◽  
pp. 133-144 ◽  
Author(s):  
Marie K. Brimnes ◽  
Laura Bonifaz ◽  
Ralph M. Steinman ◽  
Thomas M. Moran

We evaluated the proposal that during microbial infection, dendritic cells (DCs) undergo maturation and present a mixture of peptides derived from the microbe as well as harmless environmental antigens. Mice were exposed to an aerosol of endotoxin free ovalbumin (OVA) in the absence or presence of influenza virus. In its absence, OVA failed to induce B and T cell responses and even tolerized, but with influenza, OVA-specific antibodies and CD8+ cytolytic T lymphocytes developed. With or without infection, OVA was presented selectively in the draining mediastinal lymph nodes, as assessed by the comparable proliferation of infused, CD8+ and CD4+, TCR transgenic T cells. In the absence of influenza, these OVA-specific T cells produced little IL-2, IL-4, IL-10, and IFN-γ, but with infection, both CD4+ and CD8+ T cells made high levels of IL-2 and IFN-γ. The OVA plus influenza-treated mice also showed accelerated recovery to a challenge with recombinant vaccinia OVA virus. CD11c+ DCs from the mediastinal lymph nodes of infected mice selectively stimulated both OVA- and influenza-specific T cells and underwent maturation, with higher levels of MHC class II, CD80, and CD86 molecules. The relatively slow (2–3 d) kinetics of maturation correlated closely to the time at which OVA inhalation elicited specific antibodies. Therefore respiratory infection can induce DC maturation and simultaneously B and T cell immunity to an innocuous antigen inhaled concurrently.


2005 ◽  
Vol 73 (9) ◽  
pp. 5782-5788 ◽  
Author(s):  
Kyle I. Happel ◽  
Euan A. Lockhart ◽  
Carol M. Mason ◽  
Elizabeth Porretta ◽  
Elizabeth Keoshkerian ◽  
...  

ABSTRACT Interleukin-23 (IL-23) is a heterodimeric cytokine that shares IL-12 p40 but contains a unique p19 subunit similar to IL-12 p35. Previous studies indicate a greater importance for intact IL-12/23 p40 expression than IL-12 p35 for immunity against Mycobacterium tuberculosis, suggesting a role for IL-23 in host defense. The effects of IL-23 on the outcome of pulmonary infection with M. tuberculosis have not been described. Here, we show that local delivery of replication-defective adenovirus vectors encoding IL-23 (AdIL-23) greatly stimulated expression of both gamma interferon (IFN-γ) and IL-17 in lung tissues of otherwise normal mice. When given 72 h prior to infection with M. tuberculosis, AdIL-23 significantly reduced the bacterial burden at 14, 21, and 28 days. Markedly lower levels of lung inflammation were observed at 28 days than in control mice pretreated with control adenovirus (AdNull) or vehicle controls. AdIL-23 pretreatment resulted in increased numbers of CD4+ CD25+ activated T cells in lungs and draining lymph nodes compared to control groups and more CD4+ T cells bearing surface memory markers in lung lymph nodes. IL-23 gene delivery also significantly enhanced host anti-mycobacterial T-cell responses, as shown by elevated levels of IFN-γ and IL-17 secreted in vitro following restimulation with M. tuberculosis purified protein derivative. Overall, our data show that transient IL-23 gene delivery in the lung is well tolerated, and they provide the initial demonstration that this factor controls mycobacterial growth while augmenting early pulmonary T-cell immunity.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 630-630
Author(s):  
Maher K Gandhi ◽  
Rebekah M Brennan ◽  
Leesa Wockner ◽  
Pratip K Chattopadhyay ◽  
Mario Roederer ◽  
...  

Abstract In Epstein-Barr virus (EBV) classical Hodgkin lymphoma (EBV+ cHL), Hodgkin-Reed Sternberg cell antigen presentation is intact, with viral expression restricted to sub-dominant latent-antigens including LMP1/2A. Large epidemiological studies have reported differential HLA-class I (HLA-I) susceptibility to EBV+ cHL. The functional basis for these observations is unknown. HLA-I molecules present viral peptides for recognition by CD8+ T-cells, and it may be that the relative risk of developing EBV+ cHL is due to HLA-I alleles influencing the magnitude of CD8+ T-cell immunity against relevant EBV-specific antigens. However this remains speculative, with immunological evidence lacking. Several non-HLA-I linked genetic susceptibility loci have been identified, and HLA-I associations may simply represent markers for genes of diverse functions that are in linkage disequilibrium to the HLA-I region. We undertook an Australasian Leukaemia and Lymphoma Group study to address this fundamental question, utilizing 4 distinct but complimentary experimental approaches. 1. 9 EBV+ cHL and 11 EBV-ve cHL pre-therapy PBMC samples were tested for ex-vivo IFNγ, TNFα and CD107a CD8+ T-cell immunity, using overlapping LMP1 and LMP2A peptide pools. The non-HRS expressed EBV-lytic protein BZLF1 was a control. Highly stringent FACS gating was used to maximize specificity. Results were interrogated using Profile and SPICE analysis. Interestingly IFNγ, TNFα and CD107 CD8+ T-cell responses in HLA-A*02 EBV+ cHL (but not EBV-ve cHL) patients were greater than non-HLA-A*02 (LMP1 p=0.002; LMP2A p=0.03; combined LMP1/LMP2A p=0.005), whereas BZLF1 was equivalent, indicating that HLA-I provides differential CD8+ T-cell immunity against relevant EBV-latent antigens in EBV+ cHL but not EBV-ve cHL. 2. However, up to 4 different HLA-A/B molecules can potentially present relevant EBV-derived epitopes in each individual, adding a confounding layer of complexity to single allele-based effects. To overcome this and enhance sensitivity, we used the mutant HLA-I 721.221 cell-line (pulsed with LMP2A), transfected with either HLA-A*01, HLA-A*02, HLA-A*03 or HLA-B*08 alleles, as antigen presenting cells to in-vitro expand LMP2A-specific CD8+ T-cells from HLA-A*02 heterozygotes. This found ∼90% of the HLA-I LMP2A response was restricted through HLA-A*02. 3. In contrast to EBV+ cHL, in EBV-post-transplant lymphoproliferative disorders (EBV+ PTLD) the immunogenic EBNA3A/3B/3C latent-antigens are expressed. We compared HLA-I associations in 110 cHL (35% EBV+ cHL) to 153 PTLD (63% EBV+ PTLD) patients. Using Bonferoni corrected statistics, we confirmed that HLA-A*02 and HLA-A*01 homozygotes had lower and higher susceptibility to EBV+ cHL respectively, and that HLA-B*37 was positively associated. Notably, no HLA-I associations with EBV+ PTLD were found. 4. To investigate the impact of HLA-I on the hierarchy of CD8+ T-cell immunity to sub-dominant (LMP1/2A) and immune-dominant (EBNA3A/3B/3C) EBV-latent proteins, we analysed the diversity of HLA-class I restricted T-cells in 30 healthy EBV+ participants. To supplement 30 ‘defined' (i.e. validated) HLA-I EBV-latent antigen epitopes and expand HLA-I coverage, we identified 31 ‘SYFPEITHI' bioinformatically ‘predicted' peptide epitopes for HLA-A*01, HLA-A*03 or HLA-B*37 restricted EBV-latent antigens. All SYFPEITHI scores were ≥21, and thermal stability circular dichroism analysis (HLA-A*01) or MHC stabilization assays on T2 cells (HLA-A*03) confirmed peptide binding to HLA-I. Ex-vivo CD107 CD8+ T-cell assays for the 61 peptides, found that sub-dominant LMP1/2A-specific peptide responses were largely confined to HLA-A*02 (Fig 1A), whilst immuno-dominant CD8+ T-cell responses were stimulated by peptides presented by numerous HLA-I alleles (Fig 1B). These data combined illustrate that differential HLA-I-associated susceptibility to EBV+ cHL reflects altered EBV latent antigen-specific CD8+ T-cell immune hierarchies. For lymphomas expressing a restricted set of poorly immunogenic proteins, even modest CD8+ T-cell responses against relevant tumor-associated proteins confer protection, with broad implications for EBV-vaccine design. Studies are required to determine if similar mechanisms are applicable to non-lymphoid EBV+ malignancies with restricted latency such as undifferentiated nasopharngeal carcinoma. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 8 ◽  
Author(s):  
Ai Fukaya-Shiba ◽  
Kouhei Otsuka ◽  
Hajime Sasaki ◽  
Mayumi Shikano ◽  
Rika Wakao

Background: The mission of medicines regulatory agencies is to ensure the timely access of innovative products for patients to improve public health. Thus, regulators should foresee evolving technologies and build expertise prior to reviewing innovative products. Novel modalities and new classes of therapeutics in biological or cell-based products represent a regulatory challenge because of knowledge gaps, as exemplified by the unexpected cytokine release syndrome in the first-in-human clinical trial of the CD28 super-agonist. Meanwhile, recent treatments harnessing T cell co-signaling pathways provide an opportunity for investigation. Therefore, this study aimed to systematically identify and evaluate novel modalities for T cell immunity to assess the need for regulatory guidance.Methods: A PubMed search was carried out using the query, “immun* AND t lymph*” to select publications. Subsequently, a citation network was created, followed by clustering and text mining to identify the modalities and classes of therapeutics under development.Results and Discussion: Analysis of the top 20 clusters revealed research domains characterized by keywords such as immune checkpoint antibody, chimeric antigen receptor (CAR)-T cells, microbiota, exosome, regulatory T cells, unconventional T cells, and vaccines. After reviewing the pharmacological concepts, clinical trial information, and available guidance, we presented a perspective on the future development of guidance for these domains.Conclusion: Bibliometric analyses identified a set of innovative modalities targeted for drug development with which regulatory guidance is going to catch up. This strategy could help in the successful development of upcoming modalities to ensure readiness for clinical application as part of horizon scanning.


2000 ◽  
Vol 68 (11) ◽  
pp. 6223-6232
Author(s):  
Magali Moretto ◽  
Lori Casciotti ◽  
Brigit Durell ◽  
Imtiaz A. Khan

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1041-1041
Author(s):  
Emily R Summerbell ◽  
Cynthia R. Giver ◽  
Sravanti Rangaraju ◽  
Katarzyna Anna Darlak ◽  
Edmund K. Waller

Abstract Introduction Vasoactive intestinal peptide (VIP) is a neuropeptide hormone that suppresses Th1 immunity and inhibits antiviral immunity. Decreased Th1 immunity is problematic for allogeneic bone marrow transplant (allo-BMT) patients requiring T-cell immunity against blood cancers (Graft-versus-Tumor) and against secondary infections such as CMV. VIPhyb, a modified VIP peptide, is a VIP receptor antagonist that decreases VIP signaling. VIP-knockout mice and mice treated with VIPhyb after allo-BMT are known to have better antiviral immunity and survival after CMV infection without increasing GvHD (Li et al. PLoS One. 2013 May 27;8(5):e63381) (Li et al. Blood. 2013 Mar 21;121(12):2347-51.), thus making VIPhyb of interest for pharmacological use in humans to improve the efficacy of allo-BMT The effects of VIPhyb on T-cell immunity are not yet fully profiled. This study aimed to analyze the effects of VIPhyb on CD4+ and CD8+ T-cell proliferation and activation in order to better understand the mechanistic implications of VIP inhibition on T-cell adaptive immunity. This study also aimed to show that mixed lymphocyte reactions (MLRs), an in vitro allo-BMT model, could be used to provide rapid and reliable results that are consistent with in vivo data. It was hypothesized that VIPhyb would increase T-cell immunity as profiled by: increased T-cell proliferation, CD69 and PD1 co-upregulation in early T-cell activation, and PD1 downregulation in T-cells after initial activation. Methods Splenocytes from two histoincompatible mice were cultured together at 37°C in a 1:1 ratio in a one-way MLR. BALB/c splenocytes (stimulators) were irradiated at 20Gy, and Pepboy splenocytes (responders) were labeled with CFSE to trace proliferation. VIPhyb was added daily to the cell cultures in doses of 0.1μM, 0.3μM, 1μM, or 3μM. Treatment groups were compared to a PBS control. Proliferation, CD69, and PD1 were assessed by flow cytometry on the BD FACSAria. All results are shown as mean ± SEM (n=3). One-way ANOVA tests with Dunnett post-tests were calculated using Prism software. *p < 0.05; **p < 0.01; ***p < 0.001 Results VIPhyb increased CD4+ and CD8+ T-cell proliferation: 3, 5, and 7 days after initiating a one-way MLR, CFSE expression of Pepboy responder T-cells was assessed using flow cytometry (Figure 1). As the VIPhyb dose increased, the percentage of initial splenocytes that underwent proliferation increased in both CD4+ and CD8+ T-cells. VIPhyb increased early T-cell CD69 expression and abrogated later PD1 upregulation in CD8+ T-cells: 3, 5, and 7 days after initiating a one-way MLR, expression levels of CD69 and PD1 on Pepboy responder T-cells were assessed by flow cytometry. Significant upregulation of CD69 on CD4+ and CD8+ T-cells on day 3 occurred with increasing VIPhyb doses (Figures 2A and 2B). PD1 was co-upregulated with CD69 during early activation, and VIPhyb significantly decreased PD1 expression on CD8+ T-cells on days 5 and 7 (Figures 2C and 2D). Conclusions VIPhyb increased T-cell proliferation; CD8+ T-cells were affected more significantly. VIPhyb increased early co-upregulation of CD69 and PD1 in all T-cells and significantly decreased later CD8+ T-cell PD1 expression, indicating that VIPhyb increases T-cell activation. We hypothesize that the decreased PD1 expression will be critical for understanding the pathways involved in VIP inhibition. Importantly, since it has been shown in vivo that VIPhyb does not increase GvHD, then it can be assumed that the VIPhyb-induced T-cell proliferation and activation will increase GvL and adaptive immunity without increasing alloreactivity. Notably, these results are consistent with published in vivo data, which demonstrates that the MLR can be used as a faster method of analyzing pharmacological compounds than in vivo experiments. Given these results, VIPhyb is still of interest as a potential therapy for allo-BMT patients. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document