LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 4 of total 4

Search options

  1. Article ; Online: Radiation resistance in head and neck squamous cell carcinoma: dire need for an appropriate sensitizer.

    Hutchinson, Marsha-Kay N D / Mierzwa, Michelle / D'Silva, Nisha J

    Oncogene

    2020  Volume 39, Issue 18, Page(s) 3638–3649

    Abstract: Radiation is a significant treatment for patients with head and neck cancer. Despite advances to improve treatment, many tumors acquire radiation resistance resulting in poor survival. Radiation kills cancer cells by inducing DNA double-strand breaks. ... ...

    Abstract Radiation is a significant treatment for patients with head and neck cancer. Despite advances to improve treatment, many tumors acquire radiation resistance resulting in poor survival. Radiation kills cancer cells by inducing DNA double-strand breaks. Therefore, radiation resistance is enhanced by efficient repair of damaged DNA. Head and neck cancers overexpress EGFR and have a high frequency of p53 mutations, both of which enhance DNA repair. This review discusses the clinical criteria for radiation resistance in patients with head and neck cancer and summarizes how cancer cells evade radiation-mediated apoptosis by p53- and epidermal growth factor receptor (EGFR)-mediated DNA repair. In addition, we explore the role of cancer stem cells in promoting radiation resistance, and how the abscopal effect provides rationale for combination strategies with immunotherapy.
    MeSH term(s) Apoptosis/radiation effects ; Cell Proliferation/radiation effects ; DNA Breaks, Double-Stranded/radiation effects ; DNA Damage/radiation effects ; DNA Repair/radiation effects ; ErbB Receptors/genetics ; Gene Expression Regulation, Neoplastic/radiation effects ; Humans ; Radiation Tolerance/genetics ; Squamous Cell Carcinoma of Head and Neck/genetics ; Squamous Cell Carcinoma of Head and Neck/pathology ; Squamous Cell Carcinoma of Head and Neck/radiotherapy ; Tumor Suppressor Protein p53/genetics
    Chemical Substances TP53 protein, human ; Tumor Suppressor Protein p53 ; EGFR protein, human (EC 2.7.10.1) ; ErbB Receptors (EC 2.7.10.1)
    Language English
    Publishing date 2020-03-10
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Review
    ZDB-ID 639046-8
    ISSN 1476-5594 ; 0950-9232
    ISSN (online) 1476-5594
    ISSN 0950-9232
    DOI 10.1038/s41388-020-1250-3
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Reversing Epigenetic Gene Silencing to Overcome Immune Evasion in CNS Malignancies.

    Ratnam, Nivedita M / Sonnemann, Heather M / Frederico, Stephen C / Chen, Huanwen / Hutchinson, Marsha-Kay N D / Dowdy, Tyrone / Reid, Caitlin M / Jung, Jinkyu / Zhang, Wei / Song, Hua / Zhang, Meili / Davis, Dionne / Larion, Mioara / Giles, Amber J / Gilbert, Mark R

    Frontiers in oncology

    2021  Volume 11, Page(s) 719091

    Abstract: Glioblastoma (GBM) is an aggressive brain malignancy with a dismal prognosis. With emerging evidence to disprove brain-immune privilege, there has been much interest in examining immunotherapy strategies to treat central nervous system (CNS) cancers. ... ...

    Abstract Glioblastoma (GBM) is an aggressive brain malignancy with a dismal prognosis. With emerging evidence to disprove brain-immune privilege, there has been much interest in examining immunotherapy strategies to treat central nervous system (CNS) cancers. Unfortunately, the limited success of clinical studies investigating immunotherapy regimens, has led to questions about the suitability of immunotherapy for these cancers. Inadequate inherent populations of tumor infiltrating lymphocytes (TILs) and limited trafficking of systemic, circulating T cells into the CNS likely contribute to the poor response to immunotherapy. This paucity of TILs is in concert with the finding of epigenetic silencing of genes that promote immune cell movement (chemotaxis) to the tumor. In this study we evaluated the ability of GSK126, a blood-brain barrier (BBB) permeable small molecule inhibitor of EZH2, to reverse GBM immune evasion by epigenetic suppression of T cell chemotaxis. We also evaluated the
    Language English
    Publishing date 2021-07-15
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2649216-7
    ISSN 2234-943X
    ISSN 2234-943X
    DOI 10.3389/fonc.2021.719091
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Phosphorylation of TRIP13 at Y56 induces radiation resistance but sensitizes head and neck cancer to cetuximab.

    Banerjee, Rajat / Liu, Min / Bellile, Emily / Schmitd, Ligia B / Goto, Mitsuo / Hutchinson, Marsha-Kay N D / Singh, Priyanka / Zhang, Shuang / Damodaran, Dilna P V / Nyati, Mukesh K / Spector, Matthew E / Ward, Brent / Wolf, Gregory / Casper, Keith / Mierzwa, Michelle / D'Silva, Nisha J

    Molecular therapy : the journal of the American Society of Gene Therapy

    2021  Volume 30, Issue 1, Page(s) 468–484

    Abstract: Radiation therapy, a mainstay of treatment for head and neck cancer, is not always curative due to the development of treatment resistance; additionally, multi-institutional trials have questioned the efficacy of concurrent radiation with cetuximab, the ... ...

    Abstract Radiation therapy, a mainstay of treatment for head and neck cancer, is not always curative due to the development of treatment resistance; additionally, multi-institutional trials have questioned the efficacy of concurrent radiation with cetuximab, the epidermal growth factor receptor (EGFR) inhibitor. We unraveled a mechanism for radiation resistance; that is, radiation induces EGFR, which phosphorylates TRIP13 (thyroid hormone receptor interactor 13) on tyrosine 56. Phosphorylated (phospho-)TRIP13 promotes non-homologous end joining (NHEJ) repair to induce radiation resistance. NHEJ is the main repair pathway for radiation-induced DNA damage. Tumors expressing high TRIP13 do not respond to radiation but are sensitive to cetuximab or cetuximab combined with radiation. Suppression of phosphorylation of TRIP13 at Y56 abrogates these effects. These findings show that EGFR-mediated phosphorylation of TRIP13 at Y56 is a vital mechanism of radiation resistance. Notably, TRIP13-pY56 could be used to predict the response to radiation or cetuximab and could be explored as an actionable target.
    MeSH term(s) ATPases Associated with Diverse Cellular Activities/metabolism ; Cell Cycle Proteins/metabolism ; Cell Line, Tumor ; Cetuximab/metabolism ; Cetuximab/pharmacology ; DNA End-Joining Repair ; Head and Neck Neoplasms/drug therapy ; Head and Neck Neoplasms/genetics ; Head and Neck Neoplasms/radiotherapy ; Humans ; Phosphorylation
    Chemical Substances Cell Cycle Proteins ; ATPases Associated with Diverse Cellular Activities (EC 3.6.4.-) ; TRIP13 protein, human (EC 3.6.4.-) ; Cetuximab (PQX0D8J21J)
    Language English
    Publishing date 2021-06-08
    Publishing country United States
    Document type Journal Article ; Research Support, N.I.H., Extramural
    ZDB-ID 2010592-7
    ISSN 1525-0024 ; 1525-0016
    ISSN (online) 1525-0024
    ISSN 1525-0016
    DOI 10.1016/j.ymthe.2021.06.009
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Dexamethasone-induced immunosuppression: mechanisms and implications for immunotherapy.

    Giles, Amber J / Hutchinson, Marsha-Kay N D / Sonnemann, Heather M / Jung, Jinkyu / Fecci, Peter E / Ratnam, Nivedita M / Zhang, Wei / Song, Hua / Bailey, Rolanda / Davis, Dionne / Reid, Caitlin M / Park, Deric M / Gilbert, Mark R

    Journal for immunotherapy of cancer

    2018  Volume 6, Issue 1, Page(s) 51

    Abstract: Background: Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is ... ...

    Abstract Background: Corticosteroids are routinely utilized to alleviate edema in patients with intracranial lesions and are first-line agents to combat immune-related adverse events (irAEs) that arise with immune checkpoint blockade treatment. However, it is not known if or when corticosteroids can be administered without abrogating the efforts of immunotherapy. The purpose of this study was to evaluate the impact of dexamethasone on lymphocyte activation and proliferation during checkpoint blockade to provide guidance for corticosteroid use while immunotherapy is being implemented as a cancer treatment.
    Methods: Lymphocyte proliferation, differentiation, and cytokine production were evaluated during dexamethasone exposure. Human T cells were stimulated through CD3 ligation and co-stimulated either directly by CD28 ligation or by providing CD80, a shared ligand for CD28 and CTLA-4. CTLA-4 signaling was inhibited by antibody blockade using ipilimumab which has been approved for the treatment of several solid tumors. The in vivo effects of dexamethasone during checkpoint blockade were evaluated using the GL261 syngeneic mouse intracranial model, and immune populations were profiled by flow cytometry.
    Results: Dexamethasone upregulated CTLA-4 mRNA and protein in CD4 and CD8 T cells and blocked CD28-mediated cell cycle entry and differentiation. Naïve T cells were most sensitive, leading to a decrease of the development of more differentiated subsets. Resistance to dexamethasone was conferred by blocking CTLA-4 or providing strong CD28 co-stimulation prior to dexamethasone exposure. CTLA-4 blockade increased IFNγ expression, but not IL-2, in stimulated human peripheral blood T cells exposed to dexamethasone. Finally, we found that CTLA-4 blockade partially rescued T cell numbers in mice bearing intracranial gliomas. CTLA-4 blockade was associated with increased IFNγ-producing tumor-infiltrating T cells and extended survival of dexamethasone-treated mice.
    Conclusions: Dexamethasone-mediated T cell suppression diminishes naïve T cell proliferation and differentiation by attenuating the CD28 co-stimulatory pathway. However, CTLA-4, but not PD-1 blockade can partially prevent some of the inhibitory effects of dexamethasone on the immune response.
    MeSH term(s) Animals ; Dexamethasone/pharmacology ; Dexamethasone/therapeutic use ; Disease Models, Animal ; Female ; Humans ; Immunosuppression/methods ; Immunotherapy/methods ; Mice
    Chemical Substances Dexamethasone (7S5I7G3JQL)
    Language English
    Publishing date 2018-06-11
    Publishing country England
    Document type Journal Article ; Research Support, N.I.H., Intramural
    ZDB-ID 2719863-7
    ISSN 2051-1426 ; 2051-1426
    ISSN (online) 2051-1426
    ISSN 2051-1426
    DOI 10.1186/s40425-018-0371-5
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top