LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 3 of total 3

Search options

  1. Article ; Online: TREM1 activation of myeloid cells promotes antitumor immunity.

    Juric, Vladislava / Mayes, Erin / Binnewies, Mikhail / Lee, Tian / Canaday, Pamela / Pollack, Joshua L / Rudolph, Joshua / Du, Xiaoyan / Liu, Victoria M / Dash, Subhadra / Palmer, Rachael / Jahchan, Nadine S / Ramoth, Åsa Johanna / Lacayo, Sergio / Mankikar, Shilpa / Norng, Manith / Brassell, Chris / Pal, Aritra / Chan, Christopher /
    Lu, Erick / Sriram, Venkataraman / Streuli, Michel / Krummel, Matthew F / Baker, Kevin P / Liang, Linda

    Science translational medicine

    2023  Volume 15, Issue 711, Page(s) eadd9990

    Abstract: Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor ...

    Abstract Myeloid cells in the tumor microenvironment (TME) can exist in immunosuppressive and immunostimulatory states that impede or promote antitumor immunity, respectively. Blocking suppressive myeloid cells or increasing stimulatory cells to enhance antitumor immune responses is an area of interest for therapeutic intervention. Triggering receptor expressed on myeloid cells-1 (TREM1) is a proinflammatory receptor that amplifies immune responses. TREM1 is expressed on neutrophils, subsets of monocytes and tissue macrophages, and suppressive myeloid populations in the TME, including tumor-associated neutrophils, monocytes, and tumor-associated macrophages. Depletion or inhibition of immunosuppressive myeloid cells, or stimulation by TREM1-mediated inflammatory signaling, could be used to promote an immunostimulatory TME. We developed PY159, an afucosylated humanized anti-TREM1 monoclonal antibody with enhanced FcγR binding. PY159 is a TREM1 agonist that induces signaling, leading to up-regulation of costimulatory molecules on monocytes and macrophages, production of proinflammatory cytokines and chemokines, and enhancement of T cell activation in vitro. An antibody against mouse TREM1, PY159m, promoted antitumor efficacy in syngeneic mouse tumor models. These results suggest that PY159-mediated agonism of TREM1 on tumoral myeloid cells can promote a proinflammatory TME and offer a promising strategy for immunotherapy.
    MeSH term(s) Animals ; Mice ; Antibodies, Monoclonal/pharmacology ; Antibodies, Monoclonal/therapeutic use ; Disease Models, Animal ; Immunosuppressive Agents ; Macrophages ; Monocytes ; Myeloid Cells ; Triggering Receptor Expressed on Myeloid Cells-1
    Chemical Substances Antibodies, Monoclonal ; Immunosuppressive Agents ; Triggering Receptor Expressed on Myeloid Cells-1 ; TREM1 protein, mouse
    Language English
    Publishing date 2023-08-30
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2518854-9
    ISSN 1946-6242 ; 1946-6234
    ISSN (online) 1946-6242
    ISSN 1946-6234
    DOI 10.1126/scitranslmed.add9990
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Targeting TREM2 on tumor-associated macrophages enhances immunotherapy.

    Binnewies, Mikhail / Pollack, Joshua L / Rudolph, Joshua / Dash, Subhadra / Abushawish, Marwan / Lee, Tian / Jahchan, Nadine S / Canaday, Pamela / Lu, Erick / Norng, Manith / Mankikar, Shilpa / Liu, Victoria M / Du, Xiaoyan / Chen, Amanda / Mehta, Ranna / Palmer, Rachael / Juric, Vladislava / Liang, Linda / Baker, Kevin P /
    Reyno, Leonard / Krummel, Matthew F / Streuli, Michel / Sriram, Venkataraman

    Cell reports

    2021  Volume 37, Issue 3, Page(s) 109844

    Abstract: Converting checkpoint inhibitor (CPI)-resistant individuals to being responsive requires identifying suppressive mechanisms. We identify ... ...

    Abstract Converting checkpoint inhibitor (CPI)-resistant individuals to being responsive requires identifying suppressive mechanisms. We identify TREM2
    MeSH term(s) Animals ; Antineoplastic Agents, Immunological/pharmacology ; Antineoplastic Combined Chemotherapy Protocols/pharmacology ; CD8-Positive T-Lymphocytes/drug effects ; CD8-Positive T-Lymphocytes/immunology ; CD8-Positive T-Lymphocytes/metabolism ; Cell Line, Tumor ; Coculture Techniques ; Drug Resistance, Neoplasm ; Female ; HEK293 Cells ; Humans ; Immune Checkpoint Inhibitors/pharmacology ; Lymphocyte Activation/drug effects ; Lymphocytes, Tumor-Infiltrating/drug effects ; Lymphocytes, Tumor-Infiltrating/immunology ; Lymphocytes, Tumor-Infiltrating/metabolism ; Membrane Glycoproteins/antagonists & inhibitors ; Membrane Glycoproteins/metabolism ; Mice, Inbred BALB C ; Mice, Inbred C57BL ; Neoplasms/drug therapy ; Neoplasms/immunology ; Neoplasms/metabolism ; Neoplasms/pathology ; Programmed Cell Death 1 Receptor/antagonists & inhibitors ; Programmed Cell Death 1 Receptor/immunology ; Programmed Cell Death 1 Receptor/metabolism ; Receptors, Immunologic/antagonists & inhibitors ; Receptors, Immunologic/metabolism ; Signal Transduction ; Tumor Cells, Cultured ; Tumor Microenvironment ; Tumor-Associated Macrophages/drug effects ; Tumor-Associated Macrophages/immunology ; Tumor-Associated Macrophages/metabolism ; Mice
    Chemical Substances Antineoplastic Agents, Immunological ; Immune Checkpoint Inhibitors ; Membrane Glycoproteins ; PDCD1 protein, human ; Pdcd1 protein, mouse ; Programmed Cell Death 1 Receptor ; Receptors, Immunologic ; TREM2 protein, human ; Trem2 protein, mouse
    Language English
    Publishing date 2021-10-19
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2649101-1
    ISSN 2211-1247 ; 2211-1247
    ISSN (online) 2211-1247
    ISSN 2211-1247
    DOI 10.1016/j.celrep.2021.109844
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Structural basis for cancer immunotherapy by the first-in-class checkpoint inhibitor ipilimumab.

    Ramagopal, Udupi A / Liu, Weifeng / Garrett-Thomson, Sarah C / Bonanno, Jeffrey B / Yan, Qingrong / Srinivasan, Mohan / Wong, Susan C / Bell, Alasdair / Mankikar, Shilpa / Rangan, Vangipuram S / Deshpande, Shrikant / Korman, Alan J / Almo, Steven C

    Proceedings of the National Academy of Sciences of the United States of America

    2017  Volume 114, Issue 21, Page(s) E4223–E4232

    Abstract: Rational modulation of the immune response with biologics represents one of the most promising and active areas for the realization of new therapeutic strategies. In particular, the use of function blocking monoclonal antibodies targeting checkpoint ... ...

    Abstract Rational modulation of the immune response with biologics represents one of the most promising and active areas for the realization of new therapeutic strategies. In particular, the use of function blocking monoclonal antibodies targeting checkpoint inhibitors such as CTLA-4 and PD-1 have proven to be highly effective for the systemic activation of the human immune system to treat a wide range of cancers. Ipilimumab is a fully human antibody targeting CTLA-4 that received FDA approval for the treatment of metastatic melanoma in 2011. Ipilimumab is the first-in-class immunotherapeutic for blockade of CTLA-4 and significantly benefits overall survival of patients with metastatic melanoma. Understanding the chemical and physical determinants recognized by these mAbs provides direct insight into the mechanisms of pathway blockade, the organization of the antigen-antibody complexes at the cell surface, and opportunities to further engineer affinity and selectivity. Here, we report the 3.0 Å resolution X-ray crystal structure of the complex formed by ipilimumab with its human CTLA-4 target. This structure reveals that ipilimumab contacts the front β-sheet of CTLA-4 and intersects with the CTLA-4:Β7 recognition surface, indicating that direct steric overlap between ipilimumab and the B7 ligands is a major mechanistic contributor to ipilimumab function. The crystallographically observed binding interface was confirmed by a comprehensive cell-based binding assay against a library of CTLA-4 mutants and by direct biochemical approaches. This structure also highlights determinants responsible for the selectivity exhibited by ipilimumab toward CTLA-4 relative to the homologous and functionally related CD28.
    Language English
    Publishing date 2017-05-23
    Publishing country United States
    Document type Journal Article
    ZDB-ID 209104-5
    ISSN 1091-6490 ; 0027-8424
    ISSN (online) 1091-6490
    ISSN 0027-8424
    DOI 10.1073/pnas.1617941114
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top