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: Journal of Clinical Investigation, American Society for Clinical Investigation, Vol. 124, No. 1 ( 2014-1-2), p. 198-208
    Type of Medium: Online Resource
    ISSN: 0021-9738
    Language: English
    Publisher: American Society for Clinical Investigation
    Publication Date: 2014
    detail.hit.zdb_id: 2018375-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2443-2443
    Abstract: Background:Harnessing the cytotoxic properties of T cells to destroy tumor cells has been the central goal of anti-cancer immunotherapy. One approach to achieve this goal has been the use adoptive cell transfer (ACT). This method involves the isolation, in vitro manipulation and re-infusion of antigen-specific T cells into cancer patients. This approach has been used to mediate durable responses in a number of malignancies. Despite its significant therapeutic potential, the routine use of ACT has been limited. This has been due, in large part, to the high cost, intricate and labor-intensive methods required to successfully generate autologous antigen-specific T cells. There is a clear need for less expensive more efficient and streamlined approaches for generation of antigen-specific T cells for use in ACT. To this end, we have developed a T cell Enrichment+Expansion strategy using paramagnetic, Nano scale artificial Antigen Presenting Cells (nano-aAPCs), which are capable of enriching rare tumor-specific T cells in a magnetic column and activating them. Nano-aAPCs, are not only more biocompatible than traditional aAPC, but by manipulating paramagnetic nanoparticle based aAPC with magnetic fields, we propose a method to quickly generate large numbers of high frequency tumor-specific T cells. Our hypothesis is that magnetic enrichment and expansion with nano-aAPC can reduce time in culture and increase frequency of antigen-specific cells, two factors that we predict will improve T cell expansion and persistence after adoptive transfer. Methods:HLA-Ig dimer was loaded with tumor antigen peptides for MART1, NY-ESO and WT1. To manufacture nano-APCs, magnetic nanoparticles (size 50-100nm) were decorated with loaded HLA-Ig (signal 1) and anti-CD28 antibody (signal 2). Peripheral blood mononuclear cells (PBMCs) were collected from normal donors and CD8+ T cells were isolated. CD8+ T cells were subsequently incubated with nano-APCs. The T cell/nano-APCs mixtures were then pass through a magnetic column. The relevant T cells were bound to the nano-APCs and therefore were attached to the column while the irrelevant T cells passed through the column. Relevant T cells were then washed off the column and cultured using standard T cell culture techniques for 7 days. Cell number and specificity were evaluated on Day 0 and day 7. Results: CD8+ T cells specific for the tumor antigens MART1, NYESO and WT1 were successfully expanded using our nano-APCs. After one week, we saw fold expansions of 〉 100-fold for all 3 antigens studied. T cell specificity increased from 〈 1% for all 3 antigens on day 0 to 27% for NY-ESO, 16% for MART1 and 4% for WT1 on day 7. These numbers are comparable, and in most cases a vast improvement, to the numbers of antigen specific cells obtained using conventional methods in 7 days (i.e. dendritic cells as APCs). Conclusions: The initial data presented here represent proof of principle evidence of the feasibility and efficacy of our proposed streamlined and cost effective enrichment and expansion approach for the generation of antigen specific T cells. Here we expanded significant numbers of CD8+ T cells targeting the tumor antigens MART1 NY-ESO and WT1 in just 7 days using our nano-APCs. While MART1 was used a model antigen to develop or system, both NY-ESO and WT1 are tumor antigens that are relevant for several hematological malignancies. Streamlining the generation of large numbers of high-frequency tumor-specific T cells in a cost effective, reproducible fashion through Enrichment+Expansion could be a powerful addition to current tumor immunotherapy protocols. Disclosures Oelke: NextImmune: Equity Ownership. Schneck:NextImmune: Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Impact Journals, LLC ; 2016
    In:  Oncotarget Vol. 7, No. 42 ( 2016-10-12), p. 68503-68512
    In: Oncotarget, Impact Journals, LLC, Vol. 7, No. 42 ( 2016-10-12), p. 68503-68512
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Clinical Cancer Research Vol. 21, No. 9 ( 2015-05-01), p. 2075-2083
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 9 ( 2015-05-01), p. 2075-2083
    Abstract: Purpose: Artificial antigen-presenting cells, aAPC, have successfully been used to stimulate antigen-specific T-cell responses in vitro as well as in vivo. Although aAPC compare favorably with autologous dendritic cells in vitro, their effect in vivo might be diminished through rapid clearance by macrophages. Therefore, to prevent uptake and minimize clearance of aAPC by macrophages, thereby increasing in vivo functionality, we investigated the efficiency of “don't eat me” three-signal aAPC compared with classical two-signal aAPC. Experimental Design: To generate “don't eat me” aAPC, CD47 was additionally immobilized onto classical aAPC (aAPCCD47+). aAPC and aAPCCD47+ were analyzed in in vitro human primary T-cell and macrophage cocultures. In vivo efficiency was compared in a NOD/SCID T-cell proliferation and a B16-SIY melanoma model. Results: This study demonstrates that aAPCCD47+ in coculture with human macrophages show a CD47 concentration–dependent inhibition of phagocytosis, whereas their ability to generate and expand antigen-specific T cells was not affected. Furthermore, aAPCCD47+-generated T cells displayed equivalent killing abilities and polyfunctionality when compared with aAPC-generated T cells. In addition, in vivo studies demonstrated an enhanced stimulatory capacity and tumor inhibition of aAPCCD47+ over normal aAPC in conjunction with diverging biodistribution in different organs. Conclusions: Our data for the first time show that aAPC functionalized with CD47 maintain their stimulatory capacity in vitro and demonstrate enhanced in vivo efficiency. Thus, these next-generation aAPCCD47+ have a unique potential to enhance the application of the aAPC technology for future immunotherapy approaches. Clin Cancer Res; 21(9); 2075–83. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    The American Association of Immunologists ; 2013
    In:  The Journal of Immunology Vol. 190, No. 1_Supplement ( 2013-05-01), p. 65.4-65.4
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 190, No. 1_Supplement ( 2013-05-01), p. 65.4-65.4
    Abstract: The ability for a single T cell to upregulate multiple effector functions, polyfunctionality, is crucial for protective immunity. Loss of polyfunctional T cells during chronic viral infections is associated with pathogen persistence and results in poor memory T cell formation and progressive T cell exhaustion. However, the molecular mechanisms determining the generation and maintenance of polyfunctional T cells remain unclear. We found that CD8+ T cell polyfunctionality can be directly regulated by antigen dose. High antigen dose stimulation induced robust T cell proliferation but, in contrast, only poorly polyfunctional T cells. Microarray analysis revealed that poorly polyfunctional T cells induced by high dose antigen had a unique transcription profile similar to exhausted T cells observed during chronic viral infections including upregulation of inhibitory receptors. However, the poorly polyfunctional T cell state was independent of inhibitory receptor signaling. Instead, biochemical and genetic experiments showed that the inhibition of MAPK/ERK pathway through upregulation of spry2, a negative regulator of the MAPK/ERK pathway, directly modulated polyfunctionality by selectively inhibiting cytokine secretion. Our findings reveal novel mechanisms controlling human T cell polyfunctionality and are directly relevant to effective vaccine development and T cell immunotherapy.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2013
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Oncotarget, Impact Journals, LLC, Vol. 9, No. 64 ( 2018-08-17), p. 32401-32401
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2018
    detail.hit.zdb_id: 2560162-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 2753-2753
    Abstract: Immunotherapy is the modulation of a patient’s immune system to treat illness. Unfortunately many T cell based attempts have failed due to the fact that existing tumor-specific T cells are mostly anergic or tolerized and ex vivo generated T cells are often already of exhausted phenotype. Therefore, investigators have developed alternative approaches including bispecific antibody technology to redirect fully functional non-tumor specific T cells to the tumor. This has been primarily accomplished through targeting CD3 which is expressed on all T cells to engage and redirect them towards a molecule that is expressed on the tumor cells. Here we present a novel nanoparticle based approach to selectively target cytotoxic T cells (CTL) and re-direct them to kill tumors, termed ATR (Antigen-specific T cell Redirectors). ATR were generated by coupling either MHC-Ig dimer or clonotypic anti-TCR antibody 1B2 to target the effector T cell population and an anti-CD19 to re-direct those to CD19+ tumor target cells onto 50-100nm nanoparticles. Flow cytometry and microscope based data confirm that the described ATR phenotype efficiently and stably stain tumor and T cells in a dose dependent manner and ATR mediate antigen-specific conjugate formation of effector T cells and tumor target cells. We further developed two clinically relevant protocols to test and optimize our ATR in vitro. First a pre-treatment approach in which the effector T cells are pre-incubated with ATR mimicking an adoptive transfer approach and second a co-culture protocol that mimics an active immunotherapy approach of direct ATR injection. Antigen-specific ATR mediated re-direction of T cells to tumor target cells was demonstrated in 51Cr-release killing assays at low E:T ratios. Variation of ATR target-cell : effector-cell targeting molecule ratio could further increase efficacy. Finally, intra tumoral ATR injection induced T cell re-direction and reduced tumor growth in a s.c. Raji/SCIDbeige treatment model. In summary this data demonstrates that ATR target and redirect antigen-specific CTL to tumor cells that would otherwise not be recognized and mediates their lysis. ATR can be used to develop new innovative immunotherapeutic approaches for all cancers that can be targeted with antibodies or antibody-like molecules. Furthermore, ATR could also be used in conjunction with virus-specific immunization to specifically increase the targeted CTL population. Ultimately, we expect ATR and their potential for clinical applications to increase our understanding of tumor immunotherapy through T cell redirection. Disclosures Oelke: NextImmune: Equity Ownership. Schneck:NextImmune: Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 2, No. S3 ( 2014-12)
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2014
    detail.hit.zdb_id: 2719863-7
    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...