LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 6 of total 6

Search options

  1. Article ; Online: The roles of TGF-β and VEGF pathways in the suppression of antitumor immunity in melanoma and other solid tumors.

    Bu, Melissa T / Chandrasekhar, Pallavi / Ding, Lizhong / Hugo, Willy

    Pharmacology & therapeutics

    2022  Volume 240, Page(s) 108211

    Abstract: Immune checkpoint blockade (ICB) has become well-known in cancer therapy, strengthening the body's antitumor immune response rather than directly targeting cancer cells. Therapies targeting immune inhibitory checkpoints, such as PD-1, PD-L1, and CTLA-4, ... ...

    Abstract Immune checkpoint blockade (ICB) has become well-known in cancer therapy, strengthening the body's antitumor immune response rather than directly targeting cancer cells. Therapies targeting immune inhibitory checkpoints, such as PD-1, PD-L1, and CTLA-4, have resulted in impressive clinical responses across different types of solid tumors. However, as with other types of cancer treatments, ICB-based immunotherapy is hampered by both innate and acquired drug resistance. We previously reported the enrichment of gene signatures associated with wound healing, epithelial-to-mesenchymal, and angiogenesis processes in the tumors of patients with innate resistance to PD-1 checkpoint antibody therapy; we termed these the Innate Anti-PD-1 Resistance Signatures (IPRES). The TGF-β and VEGFA pathways emerge as the dominant drivers of IPRES-associated processes. Here, we review these pathways' functions, their roles in immunosuppression, and the currently available therapies that target them. We also discuss recent developments in the targeting of TGF-β using a specific antibody class termed trap antibody. The application of trap antibodies opens the promise of localized targeting of the TGF-β and VEGFA pathways within the tumor microenvironment. Such specificity may offer an enhanced therapeutic window that enables suppression of the IPRES processes in the tumor microenvironment while sparing the normal homeostatic functions of TGF-β and VEGFA in healthy tissues.
    MeSH term(s) Humans ; Programmed Cell Death 1 Receptor ; Transforming Growth Factor beta ; Vascular Endothelial Growth Factor A ; Neoplasms/therapy ; Immunotherapy/methods ; Tumor Microenvironment ; Melanoma/drug therapy ; Antibodies
    Chemical Substances Programmed Cell Death 1 Receptor ; Transforming Growth Factor beta ; Vascular Endothelial Growth Factor A ; Antibodies
    Language English
    Publishing date 2022-05-14
    Publishing country England
    Document type Journal Article ; Review ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 194735-7
    ISSN 1879-016X ; 0163-7258
    ISSN (online) 1879-016X
    ISSN 0163-7258
    DOI 10.1016/j.pharmthera.2022.108211
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: A Comparison of Murine PD-1 and PD-L1 Monoclonal Antibodies.

    Bu, Melissa T / Yuan, Long / Klee, Alyssa N / Freeman, Gordon J

    Monoclonal antibodies in immunodiagnosis and immunotherapy

    2022  Volume 41, Issue 4, Page(s) 202–209

    Abstract: Blockade of the PD-L1/PD-1 pathway has proven to be a broadly effective cancer immunotherapy. FDA-approved therapeutic monoclonal antibodies (mAbs) targeting the pathway have high affinity, blocking capacity, and low antibody effector activity. A number ... ...

    Abstract Blockade of the PD-L1/PD-1 pathway has proven to be a broadly effective cancer immunotherapy. FDA-approved therapeutic monoclonal antibodies (mAbs) targeting the pathway have high affinity, blocking capacity, and low antibody effector activity. A number of rat antimouse mAbs have been used to model cancer immunotherapy in mouse models. We set forth the amino acid sequences of mAbs specific for mouse PD-1 (29F.1A12) and PD-L1 (10F.9G2) and compare their avidities, blocking capacities, biological activities, and epitope recognition with other commonly used mAbs. Further manipulation of these sequences should facilitate better modeling of immunotherapy in mouse models and the generation of novel agents.
    MeSH term(s) Animals ; Antibodies, Monoclonal ; Antineoplastic Agents, Immunological ; B7-H1 Antigen ; Immunotherapy ; Mice ; Neoplasms ; Programmed Cell Death 1 Receptor ; Rats
    Chemical Substances Antibodies, Monoclonal ; Antineoplastic Agents, Immunological ; B7-H1 Antigen ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2022-08-04
    Publishing country United States
    Document type Journal Article
    ISSN 2167-9436
    ISSN (online) 2167-9436
    DOI 10.1089/mab.2021.0068
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: TLR agonists polarize interferon responses in conjunction with dendritic cell vaccination in malignant glioma: a randomized phase II Trial.

    Everson, Richard G / Hugo, Willy / Sun, Lu / Antonios, Joseph / Lee, Alexander / Ding, Lizhong / Bu, Melissa / Khattab, Sarah / Chavez, Carolina / Billingslea-Yoon, Emma / Salazar, Andres / Ellingson, Benjamin M / Cloughesy, Timothy F / Liau, Linda M / Prins, Robert M

    Nature communications

    2024  Volume 15, Issue 1, Page(s) 3882

    Abstract: In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade ... ...

    Abstract In this randomized phase II clinical trial, we evaluated the effectiveness of adding the TLR agonists, poly-ICLC or resiquimod, to autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination in patients with newly-diagnosed or recurrent WHO Grade III-IV malignant gliomas. The primary endpoints were to assess the most effective combination of vaccine and adjuvant in order to enhance the immune potency, along with safety. The combination of ATL-DC vaccination and TLR agonist was safe and found to enhance systemic immune responses, as indicated by increased interferon gene expression and changes in immune cell activation. Specifically, PD-1 expression increases on CD4+ T-cells, while CD38 and CD39 expression are reduced on CD8+ T cells, alongside an increase in monocytes. Poly-ICLC treatment amplifies the induction of interferon-induced genes in monocytes and T lymphocytes. Patients that exhibit higher interferon response gene expression demonstrate prolonged survival and delayed disease progression. These findings suggest that combining ATL-DC with poly-ICLC can induce a polarized interferon response in circulating monocytes and CD8+ T cells, which may represent an important blood biomarker for immunotherapy in this patient population.Trial Registration: ClinicalTrials.gov Identifier: NCT01204684.
    MeSH term(s) Humans ; Dendritic Cells/immunology ; Dendritic Cells/drug effects ; Glioma/immunology ; Glioma/therapy ; Female ; Male ; Interferons ; Middle Aged ; Cancer Vaccines/immunology ; Cancer Vaccines/administration & dosage ; Cancer Vaccines/therapeutic use ; CD8-Positive T-Lymphocytes/immunology ; CD8-Positive T-Lymphocytes/drug effects ; Poly I-C/administration & dosage ; Poly I-C/pharmacology ; Adult ; Toll-Like Receptors/agonists ; Imidazoles/pharmacology ; Imidazoles/therapeutic use ; Aged ; Vaccination ; Monocytes/immunology ; Monocytes/drug effects ; Brain Neoplasms/immunology ; Brain Neoplasms/therapy ; Brain Neoplasms/drug therapy ; CD4-Positive T-Lymphocytes/immunology ; CD4-Positive T-Lymphocytes/drug effects ; Immunotherapy/methods ; Toll-Like Receptor Agonists ; Carboxymethylcellulose Sodium/analogs & derivatives ; Polylysine/analogs & derivatives
    Chemical Substances poly ICLC (7KYP9TKT70) ; Interferons (9008-11-1) ; Cancer Vaccines ; Poly I-C (O84C90HH2L) ; resiquimod (V3DMU7PVXF) ; Toll-Like Receptors ; Imidazoles ; Toll-Like Receptor Agonists ; Carboxymethylcellulose Sodium (K679OBS311) ; Polylysine (25104-18-1)
    Language English
    Publishing date 2024-05-08
    Publishing country England
    Document type Journal Article ; Clinical Trial, Phase II ; Randomized Controlled Trial ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-024-48073-y
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Antigen presentation by clonally diverse CXCR5+ B cells to CD4 and CD8 T cells is associated with durable response to immune checkpoint inhibitors.

    Ding, Lizhong / Sun, Lu / Bu, Melissa T / Zhang, Yanjun / Scott, Lauren N / Prins, Robert M / Su, Maureen A / Lechner, Melissa G / Hugo, Willy

    Frontiers in immunology

    2023  Volume 14, Page(s) 1176994

    Abstract: Introduction: Increased T cell infiltration and interferon gamma (IFNγ) pathway activation are seen in tumors of melanoma patients who respond to ICI (immune checkpoint inhibitor) or MAPK pathway inhibitor (MAPKi) therapies. Yet, the rate of durable ... ...

    Abstract Introduction: Increased T cell infiltration and interferon gamma (IFNγ) pathway activation are seen in tumors of melanoma patients who respond to ICI (immune checkpoint inhibitor) or MAPK pathway inhibitor (MAPKi) therapies. Yet, the rate of durable tumor control after ICI is almost twice that of MAPKi, suggesting that additional mechanisms may be present in patients responding to ICI therapy that are beneficial for anti-tumor immunity.
    Methods: We used transcriptional analysis and clinical outcomes from patients treated with ICI or MAPKi therapies to delineate immune mechanisms driving tumor response.
    Results: We discovered response to ICI is associated with CXCL13-driven recruitment of CXCR5+ B cells with significantly higher clonal diversity than MAPKi. Our
    Conclusions: Response to ICI, but not to MAPKi, depends on the recruitment of CXCR5+ B cells into the tumor microenvironment and their productive tumor antigen presentation to follicular helper and cytotoxic, tumor reactive T cells. Our study highlights the potential of CXCL13 and B cell based strategies to enhance the rate of durable response in melanoma patients treated with ICI.
    MeSH term(s) Humans ; Immune Checkpoint Inhibitors/pharmacology ; Immune Checkpoint Inhibitors/therapeutic use ; Antigen Presentation ; Leukocytes, Mononuclear ; CD8-Positive T-Lymphocytes ; Receptors, Antigen, B-Cell ; Melanoma/drug therapy ; Tumor Microenvironment ; Receptors, CXCR5
    Chemical Substances Immune Checkpoint Inhibitors ; Receptors, Antigen, B-Cell ; CXCR5 protein, human ; Receptors, CXCR5
    Language English
    Publishing date 2023-06-26
    Publishing country Switzerland
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2606827-8
    ISSN 1664-3224 ; 1664-3224
    ISSN (online) 1664-3224
    ISSN 1664-3224
    DOI 10.3389/fimmu.2023.1176994
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: Dendritic Cell Vaccination in Conjunction with a TLR Agonist Polarizes Interferon Immune Responses in Malignant Glioma Patients.

    Everson, Richard G / Hugo, Willy / Sun, Lu / Antonios, Joseph / Lee, Alexander / Ding, Lizhong / Bu, Melissa / Khattab, Sarah / Chavez, Carolina / Billingslea-Yoon, Emma / Salazar, Andres / Ellingson, Benjamin M / Cloughesy, Timothy F / Liau, Linda M / Prins, Robert M

    Research square

    2023  

    Abstract: Autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination is a promising immunotherapy for patients with high grade gliomas, but responses have not been demonstrated in all patients. To determine the most effective combination of autologous ... ...

    Abstract Autologous tumor lysate-pulsed dendritic cell (ATL-DC) vaccination is a promising immunotherapy for patients with high grade gliomas, but responses have not been demonstrated in all patients. To determine the most effective combination of autologous tumor lysate-pulsed DC vaccination, with or without the adjuvant toll-like receptor (TLR) agonists poly-ICLC or resiquimod, we conducted a Phase 2 clinical trial in 23 patients with newly diagnosed or recurrent WHO Grade III-IV malignant gliomas. We then performed deep, high-dimensional immune profiling of these patients to better understand how TLR agonists may influence the systemic immune responses induced by ATL-DC vaccination. Bulk RNAseq data demonstrated highly significant upregulation of type 1 and type 2 interferon gene expression selectively in patients who received adjuvant a TLR agonist together with ATL-DC. CyTOF analysis of patient peripheral blood mononuclear cells (PBMCs) showed increased expression of PD-1 on CD4+ T-cells, decreases in CD38 and CD39 on CD8+ T cells and elevated proportion of monocytes after ATL-DC + TLR agonist administration. In addition, scRNA-seq demonstrated a higher expression fold change of IFN-induced genes with poly-ICLC treatment in both peripheral blood monocytes and T lymphocytes. Patients who had higher expression of interferon response genes lived significantly longer and had longer time to progression compared to those with lower expression. The results suggest that ATL-DC in conjunction with adjuvant poly-ICLC induces a polarized interferon response in circulating monocytes and specific activation of a CD8+ T cell population, which may represent an important blood biomarker for immunotherapy in this patient population.
    Trial registration: ClinicalTrials.gov Identifier: NCT01204684.
    Language English
    Publishing date 2023-09-12
    Publishing country United States
    Document type Preprint
    DOI 10.21203/rs.3.rs-3287211/v1
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Monitoring PD-1 Phosphorylation to Evaluate PD-1 Signaling during Antitumor Immune Responses.

    Bu, Xia / Juneja, Vikram R / Reynolds, Carol G / Mahoney, Kathleen M / Bu, Melissa T / McGuire, Kathleen A / Maleri, Seth / Hua, Ping / Zhu, Baogong / Klein, Sarah R / Greenfield, Edward A / Armand, Philippe / Ritz, Jerome / Sharpe, Arlene H / Freeman, Gordon J

    Cancer immunology research

    2021  Volume 9, Issue 12, Page(s) 1465–1475

    Abstract: PD-1 expression marks activated T cells susceptible to PD-1-mediated inhibition but not whether a PD-1-mediated signal is being delivered. Molecular predictors of response to PD-1 immune checkpoint blockade (ICB) are needed. We describe a monoclonal ... ...

    Abstract PD-1 expression marks activated T cells susceptible to PD-1-mediated inhibition but not whether a PD-1-mediated signal is being delivered. Molecular predictors of response to PD-1 immune checkpoint blockade (ICB) are needed. We describe a monoclonal antibody (mAb) that detects PD-1 signaling through the detection of phosphorylation of the immunotyrosine switch motif (ITSM) in the intracellular tail of mouse and human PD-1 (phospho-PD-1). We showed PD-1
    MeSH term(s) Animals ; Antibodies, Monoclonal/pharmacology ; Antibodies, Monoclonal/therapeutic use ; Cell Line, Tumor ; Humans ; Immunity/immunology ; Immunotherapy/methods ; Mice ; Phosphorylation ; Programmed Cell Death 1 Receptor/therapeutic use ; Signal Transduction
    Chemical Substances Antibodies, Monoclonal ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2021-10-11
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2732489-8
    ISSN 2326-6074 ; 2326-6066
    ISSN (online) 2326-6074
    ISSN 2326-6066
    DOI 10.1158/2326-6066.CIR-21-0493
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top