GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 11 ( 2020-9-25)
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2020
    detail.hit.zdb_id: 2606827-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A806-A806
    Abstract: Therapeutic cancer vaccines represent an intriguing approach to cancer immunotherapy and they have been widely explored for the last decade. As opposed to standard modalities, such as surgery and chemotherapy, an effective vaccine-based immune response may provide protection against metastatic disease. Peptide based vaccines can elicit a highly targeted immune response and include a simple, fast and cost-effective production due to recent developments in solid phase peptide synthesis. Recent development within the field of COVID-19 vaccines has highlighted the use of lipid nanoparticles as an effective drug delivery system for vaccination. Incorporation of peptide antigens into engineered micro- and nanoparticles enables induction of a potent T cell response, partly attributed to prolonged and improved antigen presentation by dendritic cells after particle internalization. Peptide-based vaccines are often based on delivery of high-affinity T cell model epitopes. However, the therapeutic relevance of vaccination with low-affinity epitopes is gaining increasing support following the observation that high-affinity epitopes can promote T cell exhaustion resulting from excessive T cell receptor stimulation. Here, we characterize and evaluate a novel lipid nanoparticle (LNP) vaccine platform that is suited for delivery of both high- and low-affinity epitopes in the setting of therapeutic cancer vaccination. Methods LNPs were formulated to carry high- or low-affinity peptide epitopes from Ovalbumin (OVA) in conjunction with the TLR7 agonist 1V270. The peptides were anchored to the surface of the LNPs via a reducible DSPE-PEG2000 linker system. The therapeutic vaccine platform was evaluated in vivo both as a monotherapy and in combination with adoptive transfer of OT-I T cells in the syngeneic B16-OVA murine melanoma model. Results The LNP vaccine promotes efficient antigen-release and ensures high, continuous antigen-presentation by antigen-presenting cells. While the LNPs can be administered via multiple routes, intratumoral vaccination favors enhanced particle uptake in dendritic cells in the tumor. Formulated with either high- or low-affinity epitopes, intratumorally delivered vaccine particles promote superior tumor-infiltration of adoptively transferred T cells, which translates into potent anti-tumor efficacy in vivo. Finally, we show that vaccination with both CD8+ and CD4+ epitopes can delay tumor growth and prolong survival in an antigen-dependent manner. Conclusions This study presents a versatile and multi-purpose LNP vaccine platform that ensures effective delivery of high- and low-affinity epitopes. Intratumoral administration promotes vaccine particle uptake by intratumoral dendritic cells, which is followed by T cell infiltration and anti-tumor efficacy in vivo. Ethics Approval All animal procedures were approved by the Danish National Animal Experiments Inspectorate.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A641-A641
    Abstract: Adoptive T cell therapy (ACT) is often accompanied by supporting immunomodulatory drugs to protect T cells from the suppressive tumor microenvironment (TME) [1]. However, systemic administration of these immunomodulators can cause serious side effects and fail to distribute optimally to exert sufficient lymphocyte stimulation within the tumor and lymphoid compartments. Loading T cells with adjuvant drugs or cytokines prior to cell transfer provides a solution to this issue, showing the potential to use T cells as vehicles to carry immunomodulatory molecules to target sites [2] . SHP2 is an important hub connecting several intracellular oncogenic signaling pathways including PD-1/PD-L1, representing a notable target for cancer immunotherapy. SHP2 inhibition has been shown to elicited tumor regression by improving CD8+ T cells activity [3]. Herein we present a lipid nanoparticle system encapsulating an SHP2 inhibitor (SHP2i) that allows high T cell loading capacity and enhances their therapeutic activity. Methods Remote-loading gradients were used to achieve high encapsulation efficiency of SHP2i into the lipid nanoparticle platform. Mouse cytotoxic T cells were loaded with SHP2i, and loading efficiency and release rates from the T cells were evaluated in vitro. Flow cytometry was used to assess T cell viability, proliferation, and phenotype. In vivo biodistribution of loaded T cells was evaluated by labeling lipid nanoparticles with gadolinium and T cells with Cell-trace-marker, which were measured with ICP-MS and Flow respectively. The therapeutic anti-tumor efficacy of the loaded T cells was demonstrated on EG.7-OVA tumor-bearing mice. Results The developed formulation allowed high T cell loading efficiency of SHPi and extended-release over 5 days. Loading T cells with lipid formulated SHP2i did not compromise cell viability and proliferation and resulted in T cells retaining a central memory phenotype than unloaded counterparts. Adoptively transferred T cells loaded with lipid nanoparticles showed the same distribution and proliferation behavior as the unloaded T cells in vivo, accumulating into tumor tissue three days post cell infusion. Loaded OT.I T cells significantly improved tumor growth inhibition and overall survival than OT.I T cells alone, with 5 out of 6 mice completely tumor-free, resulting in durable long-term responders. Conclusions Loading T cells with liposomal SHP2i before ACT allowed specific and controlled delivery of immunomodulatory drugs by T cells. The loaded T cells showed improved anti-tumor efficacy. The developed lipid formulation allows the loading of a variety of immunomodulatory drugs to T cells, which serve both as a drug delivery vehicle and enhance the tumor efficacy of the transferred cells. References Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat. Rev. Immunol. 2020. p. 651–68. Combes F, Meyer E, Sanders NN. Immune cells as tumor drug delivery vehicles. J Control Release. Elsevier; 2020;327:70–87. Yuan X, Bu H, Zhou J, Yang CY, Zhang H. Recent Advances of SHP2 Inhibitors in Cancer Therapy: Current Development and Clinical Application. J Med Chem. 2020;63:11368–96. Ethics Approval The study has been approved by the Danish Animal Experiments Inspectorate with the permit number 2020-15-0201-00482. The participants gave informed consent before taking part.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 11, No. 1 ( 2021-10-05)
    Abstract: Adoptive T-cell transfer (ACT) offers a curative therapeutic option for subsets of melanoma and hematological cancer patients. To increase response rates and broaden the applicability of ACT, it is necessary to improve the post-infusion performance of the transferred T cells. The design of improved treatment strategies includes transfer of cells with a less differentiated phenotype. Such T cell subsets have high proliferative potential but require stimulatory signals in vivo to differentiate into tumor-reactive effector T cells. Thus, combination strategies are needed to support the therapeutic implementation of less differentiated T cells. Here we show that systemic delivery of tumor-associated antigens (TAAs) facilitates in vivo priming and expansion of previously non-activated T cells and enhance the cytotoxicity of activated T cells. To achieve this in vivo priming, we use flexible delivery vehicles of TAAs and a TLR7/8 agonist. Contrasting subcutaneous delivery systems, these vehicles accumulate TAAs in the spleen, thereby achieving close proximity to both cross-presenting dendritic cells and transferred T cells, resulting in robust T-cell expansion and anti-tumor reactivity. This TAA delivery platform offers a strategy to safely potentiate the post-infusion performance of T cells using low doses of antigen and TLR7/8 agonist, and thereby enhance the effect of ACT.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2615211-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 9, No. 2_Supplement ( 2021-02-01), p. PR012-PR012
    Abstract: Despite the promise of adoptive cell therapy (ACT) in the treatment of some cancer types, success against solid tumors remains elusive due to multiple immune escape pathways. T cell therapies that affect tumor microenvironment or provide adjuvant activity in addition to tumor cell lysis hold strong potential for overcoming immune escape in solid tumors. Interleukin-12 (IL-12) is a potent stimulator of innate and adaptive immune cells that holds strong potential for cancer immunotherapy, but its clinical utility has been limited by high systemic toxicities. We have previously shown that tethering an IL-12 immunocytokine to the surface of T cells prior to adoptive cell transfer (ACT) safely improves anti-tumor efficacy by promoting T cell function specifically in the tumor. Here, we demonstrate that tethered IL-12 further delivers adjuvant activity by initiating an immune cascade involving natural killer cell-mediated activation of cross-priming dendritic cells to resulting in broader anti-tumor T cell responses. In a preclinical cell therapy model tumor-specific T cells tethered with IL-12, but not the T cells alone, triggered proliferation of tumor-specific endogenous tumor infiltrating lymphocytes (TIL). This was accompanied by increased activation and infiltration of cross-presenting dendritic cells (cDC1) in tumor-draining lymph nodes (tdLN). Analyses of cDC1 subsets revealed upregulation of CD86 expression on both migratory and resident cDC1s within the tdLN, two cell types that are critical for both tumor antigen transport to tdLN and activation of naïve CD8 T cells, respectively. This activity was specific to cDC1s, as increased activation or accumulation of type 2 dendritic cells (cDC2) was not observed. Mechanistically, the IL-12 tethered T cells upregulated expression of FLT3 ligand on NK cells, a key cytokine for cDC1 recruitment and expansion. NK cell depletion ablated cDC1 tdLN infiltration and endogenous tumor infiltrating T cell expansion, indicating that recruitment and activation of endogenous tumor-specific T cells is at least partially driven by tethered IL-12-mediated activation of NK cells to recruit and expand the cross-priming dendritic cells. Together, this demonstrates that our surface tethered cytokine technology provides powerful adjuvant activity to adoptively transferred T cells and promotes activation of endogenous anti-tumor adaptive immune response, thus promoting a broad antigenic repertoire. This approach thus holds potential to help overcome challenges of antigen escape and tumor heterogeneity that limit efficacy of T cell therapies against solid tumors. This abstract is also being presented as PO078. Citation Format: Kate L. Stokes, Ditte E. Jaehger, Alvin Pratama, Holmfridur R. Halldorsdottir, Gulzar Ahmad, Jon D. Nardozzi, Katharine L. Sackton, Thomas L. Andresen, Douglas S. Jones. Adoptive transfer of T cells surface-tethered with IL-12 activates a natural killer/dendritic cell axis to promote antigen spreading for enhanced anti-tumor efficacy [abstract]. In: Abstracts: AACR Virtual Special Conference: Tumor Immunology and Immunotherapy; 2020 Oct 19-20. Philadelphia (PA): AACR; Cancer Immunol Res 2021;9(2 Suppl):Abstract nr PR012.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Science Advances, American Association for the Advancement of Science (AAAS), Vol. 8, No. 17 ( 2022-04-29)
    Abstract: Controlled surface tethering of IL-12 enhances cell therapy by boosting T cell function and repolarizing tumor immune activity.
    Type of Medium: Online Resource
    ISSN: 2375-2548
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2022
    detail.hit.zdb_id: 2810933-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Oncotarget, Impact Journals, LLC, Vol. 7, No. 29 ( 2016-07-19), p. 45730-45744
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Molecular Medicine, Springer Science and Business Media LLC, Vol. 95, No. 7 ( 2017-7), p. 779-787
    Type of Medium: Online Resource
    ISSN: 0946-2716 , 1432-1440
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2017
    detail.hit.zdb_id: 1462132-0
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancers, MDPI AG, Vol. 13, No. 24 ( 2021-12-16), p. 6326-
    Abstract: The efficacy of anti-programmedcelldeath1therapy (aPD-1), which was recently approved for basal cell carcinoma (BCC) treatment, can be enhanced by adjuvant ablative fractional laser (AFL) in syngeneic murine tumor models. In this explorative study, we aimed to assess locally applied AFL as an adjuvant to systemic aPD-1 treatment in a clinically relevant autochthonous BCC model. BCC tumors (n = 72) were induced in Ptch1+/−K14-CreER2p53fl/fl-mice (n = 34), and the mice subsequently received aPD-1 alone, AFL alone, aPD-1+AFL, or no treatment. The outcome measures included mouse survival time, tumor clearance, tumor growth rates, and tumor immune infiltration. Both aPD-1 and AFL alone significantly increased survival time relative to untreated controls (31 d and 34.5 d, respectively vs. 14 d, p = 0.0348–0.0392). Complementing aPD-1 with AFL further promoted survival (60 d, p = 0.0198 vs. aPD-1) and improved tumor clearance and growth rates. The BCCs were poorly immune infiltrated, but aPD-1 with adjuvant AFL and AFL alone induced substantial immune cell infiltration in the tumors. Similar to AFL alone, combined aPD-1 and AFL increased neutrophil counts (4-fold, p = 0.0242), the proportion of MHCII-positive neutrophils (p = 0.0121), and concordantly, CD4+ and CD8+ T-cell infiltration (p = 0.0061–0.0242). These descriptive results suggest that the anti-tumor response that is generated by aPD-1 with adjuvant AFL is potentially promoted by increased neutrophil and T-cell engraftment in tumors. In conclusion, local AFL shows substantial promise as an adjuvant to systemic aPD-1 therapy in a clinically relevant preclinical BCC model.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2021
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...