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
Filter
Material
Language
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    International Journal of High Dilution Research - IJHDR ; 2021
    In:  International Journal of High Dilution Research - ISSN 1982-6206 Vol. 17, No. 2 ( 2021-07-16), p. 34-35
    In: International Journal of High Dilution Research - ISSN 1982-6206, International Journal of High Dilution Research - IJHDR, Vol. 17, No. 2 ( 2021-07-16), p. 34-35
    Abstract: Over the past years our research group has been testing the action of highly diluted substances and tinctures on cells from the immune system. The development of new combinations was based on laboratorial tests and assays were performed independently and in a blind manner. In this abstract we present the results of experimental studies verifying the effects of a natural complex prepared according to Hahnemann’s ancient homeopathic techniques, coded as M1. The final product was a high diluted aqueous solution without color and without odor, made by a mixture of several dynamized decimal dilutions 1, 2, and 3. Here we describe two in vivo laboratory experiments: 1) metastatic subcutaneous melanoma growth model in mice treated via inhalation for 10 min, twice a day, 12 h apart for a period of 14 days; and 2) cutaneous leishmaniasis (Leishmania amazonensis strain WHOM/BR/75/Josefa) in mice footpad cushion orally treated daily, for 30 days, being 1 mL/day. All the experiments were approved by the ethics committee, in agreement with the Experimental Animal Brazilian Council and Canadian Council on Animal Care Treatment. 1) Mice treated with M1 had significantly lower tumor burden in the subcutaneous tissue than control mice. Furthermore, tumors were impaired in proliferation and tumor related angiogenesis by the inhibition of myeloid derived suppressor cells (MDSC) positive for angiotensin II type 1 receptor (AT1R). The results indicated a mechanism of action for the anti-melanoma properties of M1: the control of AT1R expression on intratumoral MDSC, subsequently leading to reduced tumor angiogenesis. And 2) after treatment with M1, the mice had rapid and effective responses against Leishmania, by altering cytokines profile, by NO increasing (p 〈 0.05), by decreasing parasitic load (p 〈 0.001), and modifying classical maturation and biogenesis of parasitophorous vacuoles (p 〈 0.001). M1 complex decreased endocytic index (p 〈 0.001), and the % of infected macrophages (p 〈 0.05), preventing the development of lesions (p 〈 0.05) caused by L. amazonensis by increasing Th1 response (p 〈 0.05). Conclusions: M1complex can be a good candidate for a complementary therapy to conventional treatments, since all the parameters observed in vivo improved. It could be an interesting clinical tool in association to a classical anti-tumor or anti-parasitic treatment, maybe resulting in better quality of life to the patients, with less toxicity
    Type of Medium: Online Resource
    ISSN: 1982-6206
    URL: Issue
    Language: Unknown
    Publisher: International Journal of High Dilution Research - IJHDR
    Publication Date: 2021
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Ethnopharmacology, Elsevier BV, Vol. 157 ( 2014-11), p. 243-250
    Type of Medium: Online Resource
    ISSN: 0378-8741
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2014
    detail.hit.zdb_id: 1491279-X
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Toxicology in Vitro, Elsevier BV, Vol. 29, No. 8 ( 2015-12), p. 2081-2088
    Type of Medium: Online Resource
    ISSN: 0887-2333
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2015
    detail.hit.zdb_id: 1501079-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Wiley ; 2014
    In:  Immunology & Cell Biology Vol. 92, No. 3 ( 2014-03), p. 237-244
    In: Immunology & Cell Biology, Wiley, Vol. 92, No. 3 ( 2014-03), p. 237-244
    Abstract: The March 2014 issue contains a Special Feature on NK cell recognition of unconventional ligands . Natural Killer (NK) cells are bone marrow derived lymphocytes capable of mediating rapid effector responses following pathogen detection in mammals. The name “Natural Killer” derives from the ability of these cells to spontaneously lyse a large range of leukemia cell lines in vitro without prior sensitization. The reviews included in this Special Feature of Immunology and Cell Biology highlight the multifaceted roles of NK cells in detecting unconventional ligands and countering a large range of pathogens and malignancies. Covering a range of topics, including the way adhesion molecules and activating receptors are central to NK cell recognition of their targets, the reviews also highlight some of the challenges that lie ahead in harnessing NK cell function and advancing NK cell immunotherapy strategies to persistent infection, cancer and bone marrow transplantation. Immunology and Cell Biology thanks the coordinator of this Special Feature – Nicholas Huntington – for his planning and input. Further background information on this important topic is available through the accompanying web focus which links to related articles from across Nature Publishing Group.
    Type of Medium: Online Resource
    ISSN: 0818-9641 , 1440-1711
    Language: English
    Publisher: Wiley
    Publication Date: 2014
    detail.hit.zdb_id: 2011707-3
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Revista Brasileira de Ortopedia, Georg Thieme Verlag KG, Vol. 49, No. 1 ( 2014-01), p. 62-68
    Type of Medium: Online Resource
    ISSN: 0102-3616
    Language: Portuguese
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2014
    detail.hit.zdb_id: 2411301-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Revista Brasileira de Ortopedia (English Edition), Georg Thieme Verlag KG, Vol. 49, No. 1 ( 2014-01), p. 62-68
    Type of Medium: Online Resource
    ISSN: 2255-4971
    Language: English
    Publisher: Georg Thieme Verlag KG
    Publication Date: 2014
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 6, No. 4 ( 2016-04-01), p. 446-459
    Abstract: CD96 has recently been shown as a negative regulator of mouse natural killer (NK)–cell activity, with Cd96−/− mice displaying hyperresponsive NK cells upon immune challenge. In this study, we have demonstrated that blocking CD96 with a monoclonal antibody inhibited experimental metastases in three different tumor models. The antimetastatic activity of anti-CD96 was dependent on NK cells, CD226 (DNAM-1), and IFNγ, but independent of activating Fc receptors. Anti-CD96 was more effective in combination with anti–CTLA-4, anti–PD-1, or doxorubicin chemotherapy. Blocking CD96 in Tigit−/− mice significantly reduced experimental and spontaneous metastases compared with its activity in wild-type mice. Co-blockade of CD96 and PD-1 potently inhibited lung metastases, with the combination increasing local NK-cell IFNγ production and infiltration. Overall, these data demonstrate that blocking CD96 is a new and complementary immunotherapeutic strategy to reduce tumor metastases. Significance: This article illustrates the antimetastatic activity and mechanism of action of an anti-CD96 antibody that inhibits the CD96–CD155 interaction and stimulates NK-cell function. Targeting host CD96 is shown to complement surgery and conventional immune checkpoint blockade. Cancer Discov; 6(4); 446–59. ©2016 AACR. This article is highlighted in the In This Issue feature, p. 331
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2607892-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Nature Immunology, Springer Science and Business Media LLC, Vol. 24, No. 5 ( 2023-05), p. 792-801
    Type of Medium: Online Resource
    ISSN: 1529-2908 , 1529-2916
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 2026412-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1591-1591
    Abstract: Cancer immunotherapies have revolutionized cancer treatment by showing clinical efficacy across multiple cancer indications. However, tumor heterogeneity and evasion from host immune cell surveillance often limit the durability and efficacy of these strategies as monotherapies. Consequently, it is becoming common practice to combine existing anticancer treatments and novel immunotherapies to maximize clinical efficacy. The pan tumor-associated antigens MICA and MICB (MICA/B) are surface proteins induced by cellular stress, often associated with tumorigenesis, and are recognized by the NK cell activating receptor NKG2D. To evade immune recognition, cancer cells often proteolytically shed the membrane distal domains of MICA/B, leading to reduced NKG2D recognition. To combat this pervasive tumor escape mechanism and create a ubiquitous cancer targeting platform, we have developed a novel CAR-iPSC-derived NK (iNK) cell that targets the conserved α3 domain of MICA/B, rendering it resistant to inhibition by shed MICA/B. To enhance effector cell function, persistence and multi-antigen capacity, further genetic editing at the iPSC stage was conducted to equip the CAR-iNK cells with a unique IL-15/IL-15 receptor fusion, the knockout of CD38 and a novel high-affinity, non-cleavable CD16 (hnCD16) to enhance antibody-dependent cellular cytotoxicity (ADCC). In this study, we evaluated the function of multiplexed engineered MICA/B CAR iNK cells (termed FT536) in combination with monoclonal antibodies (mAbs), to elicit multi-antigen targeting, and radiation therapy, to augment surface MICA/B expression. FT536 showed superior in vitro cytotoxicity and in vivo tumor control against an array of MICA/B expressing tumor lines. Furthermore, ADCC, induced in combination with cetuximab or trastuzumab, enhanced the potency of FT536 against various solid tumor lines (p & lt;0.05). To demonstrate the capability of FT536 to synergize with irradiation therapy, we utilized a panel of tumor lines, divergent in tissue origin and MICA/B expression profiles. This approach highlighted that irradiation of the SK-BR-3 tumor line, a breast adenocarcinoma that expresses low levels of surface MICA/B and high levels of EGFR, induced the upregulation of MICA/B expression (p & lt;0.05). As anticipated, FT536 exhibited enhanced, CAR-dependent cytotoxicity against irradiated SK-BR-3 cells. Ongoing work is focused on the development of in vivo models that combine FT536 with in situ tumor irradiation and mAbs in order to promote durable responses and the elimination of resistant and heterogenous cancer cells. These data demonstrate successful targeting of MICA/B positive tumors by FT536 can be augmented by mAb and radiation therapies as first-of-kind combinatorial strategies to broadly target escape-prone tumors. Citation Format: John Goulding, Robert Blum, Bryan Hancock, Moyar Ge, Brian Groff, Soheila Shirinbak, Joy Grant, Martin Hosking, Mochtar Pribadi, Yijia Pan, Hui-Yi Chui, Shohreh Sikaroodi, Lauren Fong, Janel Huffman, Wen-I Yeh, Chia-Wei Chang, Thomas Dailey, Miguel Meza, Cokey Nguyen, Lucas Ferrari de Andrade, Tom Lee, Ryan Bjordahl, Kai W. Wucherpfennig, Bahram Valamehr. FT536: Preclinical development of a novel off-the-shelf CAR-MICA/B NK cell immunotherapy combined with radiation and antibody treatments as a first-of-kind pan-cancer targeting strategy [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1591.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 10
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A126-A126
    Abstract: Chimeric antigen receptor (CAR)-T cell therapy has revolutionized cancer treatment, but it is associated with significant dose-limiting toxicities, restricted tumor targeting (limited by specific antigen expression), and, notably, a lack of multi-antigen targeting capability to mitigate tumor associated immune evasion and heterogeneity. Furthermore, dysfunctional starting material, product inconsistency, and small manufacturing lot size limits the application and on-demand availability of CAR-T cell therapy. Methods To overcome these considerable limitations, we have developed FT536, a first-of-kind, induced pluripotent stem cell (iPSC)-derived NK (iNK) cell with a novel CAR that ubiquitously targets cancer cells through canonical stress ligand recognition. We have previously reported FT536 recognizes the conserved α3 domain of the pan-tumor associated antigens MICA and MICB (MICA/B), and is derived from a renewable master iPSC line that contains multiplexed genetic edits to enhance effector cell functionality, persistence, and multi-antigen targeting capabilities via high affinity non cleavable CD16 (hnCD16) mediated antibody dependent cellular cytotoxicity (ADCC). Here we preview the nonclinical study for the investigational new drug (IND) application for FT536. Results Utilizing a manufacturing process analogous to pharmaceutical drug product development, we demonstrate FT536 can be consistently and uniformly produced with a greater than 4x10E7 fold cellular expansion per manufacturing campaign. Furthermore, FT536 can be cryopreserved at clinical scale to support off-the-shelf clinical application, with rapid product thaw and immediate patient infusion in an out-patient setting. Functional evaluation demonstrated that FT536 uniquely possesses potent and persistent antigen specific cytolytic activity against an array of solid and hematological tumor lines. Through its hnCD16 modality, FT536 can be utilized in combination with monoclonal antibodies to provide multi-antigen targeting capabilities and in conjunction with chemotherapeutics and/or radiation that augment surface MICA/B expression. In addition, directly thawed and infused FT536 demonstrated significant tumor growth inhibition in multiple solid and liquid in vivo xenograft models, in which tumor control was further enhanced in combination with a therapeutic antibody (figure 1). Finally, ongoing studies utilizing a lung adenocarcinoma model have highlighted the sustained persistence of FT536 in lung tissue up to 33 days following a single dose infusion without the need for exogenous cytokine support. Abstract 117 Figure 1 FT536 provides statistically significant in vivo anti-tumor activity which is enhanced in combination with ADCC active monoclonal antibody therapy. (A-B) FT536 significantly reduced the number of lung and liver (not shown) metastases compared to CAR negative iNK control cells in a murine metastatic melanoma model using B16-F10 cells engineered to overexpress human MICA. (C-D) FT536 alone, and in combination with Herceptin, demonstrate significant tumor growth inhibition (TGI) compared to Herceptin alone in an orthotopic xenograft model of human lung adenocarcinoma. Conclusions Collectively, these studies demonstrate that FT536 is a highly potent, multi-tumor targeting CAR-iNK cell product that is uniform in composition and can be effectively and safely used off-the-shelf for on-demand treatment of multiple solid and hematological malignancies. An IND submission is planned for 2021, with an initial Phase 1 clinical trial to follow.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...