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. 112, No. 11 ( 2008-11-16), p. 4902-4902
    Abstract: The clinical application of T-cells for adoptive immunotherapy often requires the ex vivo stimulation and expansion of antigen-specific T cells and the re-transfer of the expanded T cells into patients. One strategy to generate such T-cell products includes the use of autologous antigen-presenting cells (APCs) which vary essentially in quality and quantity. Furthermore, other strategies for the generation of APCs such as dendritic cells (DCs) are labor intensive and expensive. To circumvent these disadvantages, many investigators have focussed their research on the development of artificial APCs (aAPCs). Recent studies have demonstrated that microbeads loaded with MHC-peptide molecules in monomeric or Ig-coupled dimeric forms and co-stimulatory antibodies such as anti-CD28 and anti-4-1BB are reproducibly capable to induce high numbers of antigen-specific T-cells. In our study, we wanted to address the question if the use of further stimulatory signals in addition to CD28/4-1BB could induce higher stimulatory capacities in aAPCs. We loaded epoxy surface activated magnetic beads (DynaBeads Epoxy M-450) with HLA-B*0702/CMV_pp65 (TPRVTGGGAM) monomer and activating antibodies (AB) and/or ligands (L) against co-stimulatory molecules of the CD28 and/or the TNF-receptor (TNFR) family: namely CD28 (AB), ICOS (AB), 4-1BB (AB+L), CD27 (L), OX40 (L). The molar ratio between MHC and the co-stimulatory molecules was between 1:1 and 1:5 with a total amount of MHC-molecules of approximately 2×105 molecules/aAPC. Cell cultures were maintained as described previously 1 and analyzed weekly by staining with HLA-B*0702/CMV_pp65 (TPRVTGGGAM) tetramers and surface antibodies against the surface markers CD3, CD8, CD45RA, CCR7, CD57 and CD25. Our results demonstrate that CD28 triggering is pivotal on aAPCs besides the antigen-specific signal for an optimal expansion of CMV-specific CTL. This signal can not be replaced by ICOS or 4-1BB either alone or in combination. The addition of ICOS and/or 4-1BB to CD28 did not significantly improve the percentage of specific T-cells after 3 weeks of culture although a trend to higher cell numbers and higher specificity when combining CD28, ICOS (AB) and 4-1BB (AB) was observed. Here, longer time periods may be required to evaluate the capacity of this combination to maintain long term cultures of antigen-specific CTL. Therefore, we always applied HLA-B*0702/CMV_pp65 monomer and CD28 (AB) on the aAPCs in further experiments and supplemented these molecules by the other co-stimulatory signals as described above. Our data suggests that the phenotype (and thereby potential effector functions/in vivo survival) varies when triggering different costimulatory pathways via aAPCs. Interestingly, we could detect a higher amount of central memory T-cells (CCR7+/CD45RA−) in samples co-stimulated with the 4-1BB antibody, compared to samples treated with other co-stimulations [e.g. MHC/CD28: 6.2 % of CD8+/Tet+ vs. MHC/CD28/4-1BB (AB): 23.5 % of CD8+/Tet+]. Another difference was seen in OX40 ligand co-stimulated samples when analyzing CD57 and CD25 expression: Here, CD8+/tetramer+ T-cells had a higher content of non-activated (CD25−) T-cells with less replicative history (CD57−) than samples co-stimulated with other co-stimulations [e.g. MHC/CD28: 23 % of CD8+/Tet+ vs. MHC/CD28/OX40 (L): 45.2 % of CD8+/Tet+] . These preliminary findings suggest that the CD28 signal on aAPCs is indispensable for expansion of CMV-specific T-cells. But the phenotype and thereby in vivo functionality may be controlled by the use of defined other co-stimulatory signals like members of the CD28 and/or TNFR family. Further tests will have to define optimal amounts of MHC-molecules on aAPCs and their ratio to co-stimulatory molecules.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    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 ...
  • 4
    In: Blood, American Society of Hematology, Vol. 111, No. 7 ( 2008-04-01), p. 3546-3552
    Abstract: Several cell-based immunotherapy strategies have been developed to specifically modulate T cell–mediated immune responses. These methods frequently rely on the utilization of tolerogenic cell–based antigen-presenting cells (APCs). However, APCs are highly sensitive to cytotoxic T-cell responses, thus limiting their therapeutic capacity. Here, we describe a novel bead-based approach to modulate T-cell responses in an antigen-specific fashion. We have generated killer artificial APCs (κaAPCs) by coupling an apoptosis-inducing α-Fas (CD95) IgM mAb together with HLA-A2 Ig molecules onto beads. These κaAPCs deplete targeted antigen-specific T cells in a Fas/Fas ligand (FasL)–dependent fashion. T-cell depletion in cocultures is rapidly initiated (30 minutes), dependent on the amount of κaAPCs and independent of activation-induced cell death (AICD). κaAPCs represent a novel technology that can control T cell–mediated immune responses, and therefore has potential for use in treatment of autoimmune diseases and allograft rejection.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2008
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2011
    In:  Immunity & Ageing Vol. 8, No. 1 ( 2011-12)
    In: Immunity & Ageing, Springer Science and Business Media LLC, Vol. 8, No. 1 ( 2011-12)
    Abstract: While influenza vaccination results in protective antibodies against primary infections, clearance of infection is primarily mediated through CD8 + T cells. Studying the CD8 + T cell response to influenza epitopes is crucial in understanding the disease associated morbidity and mortality especially in at risk populations such as the elderly. We compared the CD8 + T cell response to immunodominant and subdominant influenza epitopes in HLA-A2 + control, adult donors, aged 21-42, and in geriatric donors, aged 65 and older. Results We used a novel artificial Antigen Presenting Cell (aAPC) based stimulation assay to reveal responses that could not be detected by enzyme-linked immunosorbent spot (ELISpot). 14 younger control donors and 12 geriatric donors were enrolled in this study. The mean number of influenza-specific subdominant epitopes per control donor detected by ELISpot was only 1.4 while the mean detected by aAPC assay was 3.3 (p = 0.0096). Using the aAPC assay, 92% of the control donors responded to at least one subdominant epitopes, while 71% of control donors responded to more than one subdominant influenza-specific response. 66% of geriatric donors lacked a subdominant influenza-specific response and 33% of geriatric donors responded to only 1 subdominant epitope. The difference in subdominant response between age groups is statistically significant (p = 0.0003). Conclusion Geriatric donors lacked the broad, multi-specific response to subdominant epitopes seen in the control donors. Thus, we conclude that aging leads to a decrease in the subdominant influenza-specific CTL responses which may contribute to the increased morbidity and mortality in older individuals.
    Type of Medium: Online Resource
    ISSN: 1742-4933
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2011
    detail.hit.zdb_id: 2168941-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    BMJ ; 2014
    In:  Journal for ImmunoTherapy of Cancer Vol. 2, No. Suppl 3 ( 2014), p. P4-
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 2, No. Suppl 3 ( 2014), p. P4-
    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 ...
  • 7
    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 ...
  • 8
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 26, No. 13 ( 2020-07-01), p. 3384-3396
    Abstract: Generation of antigen-specific T cells from patients with cancer employs large numbers of peripheral blood cells and/or tumor-infiltrating cells to generate antigen-presenting and effector cells commonly requiring multiple rounds of restimulation ex vivo. We used a novel paramagnetic, nanoparticle-based artificial antigen-presenting cell (nano-aAPC) that combines anti-CD28 costimulatory and human MHC class I molecules that are loaded with antigenic peptides to rapidly expand tumor antigen–specific T cells from patients with melanoma. Experimental Design: Nano-aAPC–expressing HLA-A*0201 molecules and costimulatory anti-CD28 antibody and HLA-A*0201 molecules loaded with MART-1 or gp100 class I–restricted peptides were used to stimulate CD8 T cells purified from the peripheral blood of treatment-naïve or PD-1 antibody–treated patients with stage IV melanoma. Expanded cells were restimulated with fresh peptide-pulsed nano-aAPC at day 7. Phenotype analysis and functional assays including cytokine release, cytolysis, and measurement of avidity were conducted. Results: MART-1–specific CD8 T cells rapidly expanded up to 1,000-fold by day 14 after exposure to peptide-pulsed nano-aAPC. Expanded T cells had a predominantly stem cell memory CD45RA+/CD62L+/CD95+ phenotype; expressed ICOS, PD-1, Tim3, and LAG3; and lacked CD28. Cells from patients with melanoma were polyfunctional; highly avid; expressed IL2, IFNγ, and TNFα; and exhibited cytolytic activity against tumor cell lines. They expanded 2- to 3-fold after exposure to PD-1 antibody in vivo, and expressed a highly diverse T-cell receptor V beta repertoire. Conclusions: Peptide-pulsed nano-aAPC rapidly expanded polyfunctional antigen-specific CD8 T cells with high avidity, potent lytic function, and a stem cell memory phenotype from patients with melanoma.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    Elsevier BV ; 2009
    In:  Journal of Immunological Methods Vol. 346, No. 1-2 ( 2009-07), p. 38-44
    In: Journal of Immunological Methods, Elsevier BV, Vol. 346, No. 1-2 ( 2009-07), p. 38-44
    Type of Medium: Online Resource
    ISSN: 0022-1759
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2009
    detail.hit.zdb_id: 1500495-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 69, No. 24 ( 2009-12-15), p. 9376-9384
    Abstract: The development of effective antitumor immune responses is normally constrained by low-avidity, tumor-specific CTLs that are unable to eradicate the tumor. Strategies to rescue antitumor activity of low-avidity melanoma-specific CTLs in vivo may improve immunotherapy efficacy. To boost the in vivo effectiveness of low-avidity CTLs, we immunized mice bearing lung melanoma metastases with artificial antigen-presenting cells (aAPC), made by covalently coupling pepMHC-Ig dimers and B7.1-Ig molecules to magnetic beads. aAPC treatment induced significant tumor reduction in a mouse telomerase antigen system, and complete tumor eradication in a mouse TRP-2 antigen system, when low-avidity CTLs specific for these antigens were adoptively transferred. In addition, in an in vivo treatment model of subcutaneous melanoma, aAPC injection also augmented the activity of adoptively transferred CTLs and significantly delayed tumor growth. In vivo tumor clearance due to aAPC administration correlated with in situ proliferation of the transferred CTL. In vitro studies showed that aAPC effectively stimulated cytokine release, enhanced CTL-mediated lysis, and TCR downregulation in low-avidity CTLs. Therefore, in vivo aAPC administration represents a potentially novel approach to improve cancer immunotherapy. [Cancer Res 2009;69(24):9376–84]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    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...