LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 68

Search options

  1. Article ; Online: Method of long-term, recurrent, intracerebroventricular infusion of cellular therapy in mice.

    Chen, Chao-Hsien / Curran, Michael A

    Journal of neuroscience methods

    2022  Volume 371, Page(s) 109529

    MeSH term(s) Animals ; Blood Pressure ; Infusions, Intraventricular ; Injections, Intraventricular ; Mice
    Language English
    Publishing date 2022-02-17
    Publishing country Netherlands
    Document type Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 282721-9
    ISSN 1872-678X ; 0165-0270
    ISSN (online) 1872-678X
    ISSN 0165-0270
    DOI 10.1016/j.jneumeth.2022.109529
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Preclinical Data Supporting Antitumor Activity of PD-1 Blockade.

    Curran, Michael A

    Cancer journal (Sudbury, Mass.)

    2018  Volume 24, Issue 1, Page(s) 2–6

    Abstract: Antibodies that block the PD-1 coinhibitory receptor on T cells or its primary ligand, PD-L1, have demonstrated unprecedented efficacy across a diverse array of both solid and hematologic malignancies in the clinic. These advances were built on a ... ...

    Abstract Antibodies that block the PD-1 coinhibitory receptor on T cells or its primary ligand, PD-L1, have demonstrated unprecedented efficacy across a diverse array of both solid and hematologic malignancies in the clinic. These advances were built on a foundation of murine preclinical tumor model studies, which both demonstrated the therapeutic potential of PD-1/PD-L1 antibody blockade and also provided critical insights into the cellular and molecular processes underlying their capacity to elicit immune-mediated tumor regressions. As the field of immunotherapy moves toward higher-order combinations of agents, effective utilization of murine tumor models to optimize the composition of PD-1 antibody combination therapies, as well as their dosing and scheduling, will be essential for effective clinical translation. Novel murine models bearing human tumor xenografts and engrafted human immune systems may help close the gap between preclinical and clinical immunobiology.
    MeSH term(s) Animals ; Antibodies, Monoclonal/pharmacology ; Antibodies, Monoclonal/therapeutic use ; Antineoplastic Agents/pharmacology ; Antineoplastic Agents/therapeutic use ; B7-H1 Antigen/metabolism ; Drug Evaluation, Preclinical/methods ; Humans ; Immunotherapy/methods ; Programmed Cell Death 1 Receptor/antagonists & inhibitors
    Chemical Substances Antibodies, Monoclonal ; Antineoplastic Agents ; B7-H1 Antigen ; Programmed Cell Death 1 Receptor
    Language English
    Publishing date 2018-02-22
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 2018400-1
    ISSN 1540-336X ; 1528-9117 ; 1081-4442
    ISSN (online) 1540-336X
    ISSN 1528-9117 ; 1081-4442
    DOI 10.1097/PPO.0000000000000298
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Tumor hypermetabolism confers resistance to immunotherapy.

    Liu, Arthur / Curran, Michael A

    Seminars in cancer biology

    2020  Volume 65, Page(s) 155–163

    Abstract: Advances in our understanding of tumor immune biology and development of cancer immunotherapies have led to improved outcomes for patients that suffer from aggressive cancers such as metastatic melanoma. Despite these advances, a significant proportion ... ...

    Abstract Advances in our understanding of tumor immune biology and development of cancer immunotherapies have led to improved outcomes for patients that suffer from aggressive cancers such as metastatic melanoma. Despite these advances, a significant proportion of patients still fail to benefit, and as a result, attention has shifted to understanding how cancer cells escape immune destruction. Of particular interest is the metabolic landscape of the tumor microenvironment, as recent studies have demonstrated how both competition for essential nutrients and depletion of specific amino acids can promote T cell dysfunction. Here, we will discuss the major energetic pathways engaged by both T cells and cancer cells, metabolic substrates present in the tumor microenvironment, and emerging therapeutic strategies that seek to improve T cell metabolic fitness and bolster the antitumor immune response.
    MeSH term(s) Drug Resistance, Neoplasm/immunology ; Humans ; Immunotherapy/adverse effects ; Melanoma/immunology ; Melanoma/therapy ; Mitochondrial Diseases ; Neoplasm Metastasis ; Neoplasms/immunology ; Neoplasms/therapy ; T-Lymphocytes/drug effects ; T-Lymphocytes/immunology ; Tumor Microenvironment/drug effects ; Tumor Microenvironment/immunology
    Language English
    Publishing date 2020-01-23
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 1033980-2
    ISSN 1096-3650 ; 1044-579X
    ISSN (online) 1096-3650
    ISSN 1044-579X
    DOI 10.1016/j.semcancer.2020.01.009
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Hypoxia-activated prodrug and antiangiogenic therapies cooperatively treat pancreatic cancer but elicit immunosuppressive G-MDSC infiltration.

    Liu, Arthur / Gammon, Seth T / Pisaneschi, Federica / Boda, Akash / Ager, Casey R / Piwnica-Worms, David / Hong, David S / Curran, Michael A

    JCI insight

    2024  Volume 9, Issue 1

    Abstract: We previously showed that ablation of tumor hypoxia can sensitize tumors to immune checkpoint blockade (ICB). Here, we used a Kras+/G12D TP53+/R172H Pdx1-Cre-derived (KPC-derived) model of pancreatic adenocarcinoma to examine the tumor response and ... ...

    Abstract We previously showed that ablation of tumor hypoxia can sensitize tumors to immune checkpoint blockade (ICB). Here, we used a Kras+/G12D TP53+/R172H Pdx1-Cre-derived (KPC-derived) model of pancreatic adenocarcinoma to examine the tumor response and adaptive resistance mechanisms involved in response to 2 established methods of hypoxia-reducing therapy: the hypoxia-activated prodrug TH-302 and vascular endothelial growth factor receptor 2 (VEGFR-2) blockade. The combination of both modalities normalized tumor vasculature, increased DNA damage and cell death, and delayed tumor growth. In contrast with prior cancer models, the combination did not alleviate overall tissue hypoxia or sensitize these KPC tumors to ICB therapy despite qualitative improvements to the CD8+ T cell response. Bulk tumor RNA sequencing, flow cytometry, and adoptive myeloid cell transfer suggested that treated tumor cells increased their capacity to recruit granulocytic myeloid-derived suppressor cells (G-MDSCs) through CCL9 secretion. Blockade of the CCL9/CCR1 axis could limit G-MDSC migration, and depletion of Ly6G-positive cells could sensitize tumors to the combination of TH-302, anti-VEGFR-2, and ICB. Together, these data suggest that pancreatic tumors modulate G-MDSC migration as an adaptive response to vascular normalization and that these immunosuppressive myeloid cells act in a setting of persistent hypoxia to maintain adaptive immune resistance.
    MeSH term(s) Humans ; Pancreatic Neoplasms/pathology ; Myeloid-Derived Suppressor Cells ; Adenocarcinoma/pathology ; Vascular Endothelial Growth Factor A/metabolism ; Hypoxia/metabolism
    Chemical Substances TH 302 ; Vascular Endothelial Growth Factor A
    Language English
    Publishing date 2024-01-09
    Publishing country United States
    Document type Journal Article
    ISSN 2379-3708
    ISSN (online) 2379-3708
    DOI 10.1172/jci.insight.169150
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation.

    Curran, Michael A / Glisson, Bonnie S

    Annual review of medicine

    2018  Volume 70, Page(s) 409–424

    Abstract: The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, ... ...

    Abstract The driver and passenger mutations accumulated in the process of malignant transformation offer an adequate spectrum of immune visible alterations to the cellular proteome and resulting peptidome to render these cancers targetable-and, in theory, rejectable-by the host T cell immune response. In addition, cancers often overexpress tissue-specific and developmental antigens to which immune tolerance is incomplete. Sometimes, virally transferred oncogenes drive malignant transformation and remain expressed throughout the cancer. Despite this state of antigenic sufficiency, cancer grows progressively and overcomes all efforts of the host immune system to contain it. While therapeutic cancer vaccination can mobilize high frequencies of tumor-specific T cells, these responses remain subject to intratumoral attenuation. Antibody modulation of T cell function through checkpoint blockade or costimulatory activation can restore survival, proliferation, and effector function to these tumor-infiltrating T cells and convert otherwise subtherapeutic vaccines into potentially curative cancer immunotherapeutics.
    MeSH term(s) Animals ; CTLA-4 Antigen/administration & dosage ; CTLA-4 Antigen/immunology ; Cancer Vaccines/administration & dosage ; Cancer Vaccines/immunology ; Cell Transformation, Neoplastic/immunology ; Female ; Forecasting ; Humans ; Immunologic Factors/administration & dosage ; Immunotherapy/methods ; Male ; Neoplasms/immunology ; Neoplasms/pathology ; Neoplasms/therapy ; Pancreatic Neoplasms/immunology ; Pancreatic Neoplasms/therapy ; Prostatic Neoplasms/immunology ; Prostatic Neoplasms/therapy ; Risk Assessment ; T-Lymphocytes/immunology ; Treatment Outcome
    Chemical Substances CTLA-4 Antigen ; Cancer Vaccines ; Immunologic Factors
    Language English
    Publishing date 2018-10-31
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 207930-6
    ISSN 1545-326X ; 0066-4219
    ISSN (online) 1545-326X
    ISSN 0066-4219
    DOI 10.1146/annurev-med-050217-121900
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Hypoxia Reduction Sensitizes Refractory Cancers to Immunotherapy.

    Jayaprakash, Priyamvada / Vignali, Paolo Dario Angelo / Delgoffe, Greg M / Curran, Michael A

    Annual review of medicine

    2021  Volume 73, Page(s) 251–265

    Abstract: In order to fuel their relentless expansion, cancers must expand their vasculature to augment delivery of oxygen and essential nutrients. The disordered web of irregular vessels that results, however, leaves gaps in oxygen delivery that foster tumor ... ...

    Abstract In order to fuel their relentless expansion, cancers must expand their vasculature to augment delivery of oxygen and essential nutrients. The disordered web of irregular vessels that results, however, leaves gaps in oxygen delivery that foster tumor hypoxia. At the same time, tumor cells increase their oxidative metabolism to cope with the energetic demands of proliferation, which further worsens hypoxia due to heightened oxygen consumption. In these hypoxic, nutrient-deprived environments, tumors and suppressive stroma evolve to flourish while antitumor immunity collapses due to a combination of energetic deprivation, toxic metabolites, acidification, and other suppressive signals. Reversal of cancer hypoxia thus has the potential to increase the survival and effector function of tumor-infiltrating T cells, as well as to resensitize tumors to immunotherapy. Early clinical trials combining hypoxia reduction with immune checkpoint blockade have shown promising results in treating patients with advanced, metastatic, and therapeutically refractory cancers.
    MeSH term(s) Humans ; Hypoxia/metabolism ; Hypoxia/pathology ; Hypoxia/therapy ; Immunotherapy/methods ; Neoplasms ; T-Lymphocytes ; Tumor Microenvironment
    Language English
    Publishing date 2021-10-26
    Publishing country United States
    Document type Journal Article ; Review
    ZDB-ID 207930-6
    ISSN 1545-326X ; 0066-4219
    ISSN (online) 1545-326X
    ISSN 0066-4219
    DOI 10.1146/annurev-med-060619-022830
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Identification of Nonfunctional Alternatively Spliced Isoforms of STING in Human Acute Myeloid Leukemia.

    Boda, Akash R / Liu, Arthur J / Castro-Pando, Susana / Whitfield, Benjamin T / Molldrem, Jeffrey J / Al-Atrash, Gheath / Di Francesco, Maria Emilia / Jones, Philip / Ager, Casey R / Curran, Michael A

    Cancer research communications

    2024  Volume 4, Issue 3, Page(s) 911–918

    Abstract: Lack of robust activation of Stimulator of Interferon Genes (STING) pathway and subsequent induction of type I IFN responses is considered a barrier to antitumor immunity in acute myeloid leukemia (AML). Using common human AML cell lines as in vitro ... ...

    Abstract Lack of robust activation of Stimulator of Interferon Genes (STING) pathway and subsequent induction of type I IFN responses is considered a barrier to antitumor immunity in acute myeloid leukemia (AML). Using common human AML cell lines as in vitro tools to evaluate the efficacy of novel STING agonists, we found most AML lines to be poor producers of IFNs upon exposure to extremely potent agonists, suggesting cell-intrinsic suppression of STING signaling may occur. We observed unexpected patterns of response that did not correlate with levels of STING pathway components or of known enzymes associated with resistance. To identify a genetic basis for these observations, we cloned and sequenced STING from the cDNA of human AML cell lines and found both frequent mutations and deviations from normal RNA splicing. We identified two novel spliced isoforms of STING in these lines and validated their expression in primary human AML samples. When transduced into reporter cells, these novel STING isoforms exhibited complete insensitivity to agonist stimulation. These observations identify alternative splicing as a mechanism of STING pathway suppression and suggest that most AML silences the STING pathway through direct modification rather than through engagement of external inhibitory factors.
    Significance: We find that AML acquires resistance to innate immune activation via the STING pathway through aberrant splicing of the STING transcript including two novel forms described herein that act as dominant negatives. These data broaden understanding of how cancers evolve STING resistance, and suggest that the AML tumor microenvironment, not the cancer cell, should be the target of therapeutic interventions to activate STING.
    MeSH term(s) Humans ; Protein Isoforms/genetics ; Leukemia, Myeloid, Acute/genetics ; Alternative Splicing/genetics ; Interferon Type I/genetics ; Cell Line ; Tumor Microenvironment
    Chemical Substances Protein Isoforms ; Interferon Type I
    Language English
    Publishing date 2024-03-13
    Publishing country United States
    Document type Journal Article
    ISSN 2767-9764
    ISSN (online) 2767-9764
    DOI 10.1158/2767-9764.CRC-24-0095
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article: 4-1BB Agonists: Multi-Potent Potentiators of Tumor Immunity.

    Bartkowiak, Todd / Curran, Michael A

    Frontiers in oncology

    2015  Volume 5, Page(s) 117

    Abstract: Immunotherapy is a rapidly expanding field of oncology aimed at targeting, not the tumor itself, but the immune system combating the cancerous lesion. Of the many approaches currently under study to boost anti-tumor immune responses; modulation of immune ...

    Abstract Immunotherapy is a rapidly expanding field of oncology aimed at targeting, not the tumor itself, but the immune system combating the cancerous lesion. Of the many approaches currently under study to boost anti-tumor immune responses; modulation of immune co-receptors on lymphocytes in the tumor microenvironment has thus far proven to be the most effective. Antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte antigen-4 (CTLA-4) has become the first FDA approved immune checkpoint blockade; however, tumor infiltrating lymphocytes express a diverse array of additional stimulatory and inhibitory co-receptors, which can be targeted to boost tumor immunity. Among these, the co-stimulatory receptor 4-1BB (CD137/TNFSF9) possesses an unequaled capacity for both activation and pro-inflammatory polarization of anti-tumor lymphocytes. While functional studies of 4-1BB have focused on its prominent role in augmenting cytotoxic CD8 T cells, 4-1BB can also modulate the activity of CD4 T cells, B cells, natural killer cells, monocytes, macrophages, and dendritic cells. 4-1BB's expression on both T cells and antigen presenting cells, coupled with its capacity to promote survival, expansion, and enhanced effector function of activated T cells, has made it an alluring target for tumor immunotherapy. In contrast to immune checkpoint blocking antibodies, 4-1BB agonists can both potentiate anti-tumor and anti-viral immunity, while at the same time ameliorating autoimmune disease. Despite this, 4-1BB agonists can trigger high grade liver inflammation which has slowed their clinical development. In this review, we discuss how the underlying immunobiology of 4-1BB activation suggests the potential for therapeutically synergistic combination strategies in which immune adverse events can be minimized.
    Language English
    Publishing date 2015-06-08
    Publishing country Switzerland
    Document type Journal Article ; Review
    ZDB-ID 2649216-7
    ISSN 2234-943X
    ISSN 2234-943X
    DOI 10.3389/fonc.2015.00117
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: Immune checkpoint combinations from mouse to man.

    Ai, Midan / Curran, Michael A

    Cancer immunology, immunotherapy : CII

    2015  Volume 64, Issue 7, Page(s) 885–892

    Abstract: The discovery that antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte-associated protein 4 (CTLA-4) can restore tumor immunity against many murine transplantable tumors leading to complete rejection of established cancer ... ...

    Abstract The discovery that antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte-associated protein 4 (CTLA-4) can restore tumor immunity against many murine transplantable tumors leading to complete rejection of established cancer forever changed the field of immunotherapy. In more robust murine models as well as human cancer, however, CTLA-4 blockade alone can slow tumor growth and extend patient survival, but is rarely curative. Subsequent studies have revealed a large family of T cell immune checkpoint receptors which tumors engage to shield themselves from host immunity. As with CTLA-4, blockade of one of these additional inhibitory receptors, programmed death 1, has led to remarkable therapeutic responses against tumors of multiple lineages. Checkpoint monotherapy has demonstrated that durable, immune-mediated cures of established metastatic cancers are possible, yet the percentage of patients experiencing these outcomes remains low due to both redundant mechanisms of immune suppression in the tumor and limiting toxicity associated with some therapies. Thus, extending the curative potential of immunotherapy to a larger percentage of patients with a broader spectrum of malignancies will likely require combinations of co-inhibitory blockade and co-stimulatory activation designed to peel back multiple layers of tumor immune suppression while at the same time minimizing immune-mediated toxicity. As over a dozen T cell immune checkpoints and an additional dozen more co-stimulatory receptors have now been described, the challenge before us is to identify the most advantageous combinations of these agents based on the knowledge of their underlying biology and preclinical studies in murine tumor models.
    MeSH term(s) 4-1BB Ligand/metabolism ; Animals ; Antibodies, Monoclonal/therapeutic use ; Antibodies, Monoclonal, Humanized/therapeutic use ; Antigens, Neoplasm/genetics ; Antigens, Neoplasm/immunology ; CTLA-4 Antigen/antagonists & inhibitors ; CTLA-4 Antigen/genetics ; CTLA-4 Antigen/immunology ; Humans ; Immunotherapy ; Ipilimumab ; Mice ; Neoplasms/genetics ; Neoplasms/immunology ; Neoplasms/therapy ; Programmed Cell Death 1 Receptor/antagonists & inhibitors ; T-Lymphocytes/immunology
    Chemical Substances 4-1BB Ligand ; Antibodies, Monoclonal ; Antibodies, Monoclonal, Humanized ; Antigens, Neoplasm ; CTLA-4 Antigen ; Ipilimumab ; PDCD1 protein, human ; Programmed Cell Death 1 Receptor ; pembrolizumab (DPT0O3T46P)
    Language English
    Publishing date 2015-01-03
    Publishing country Germany
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 195342-4
    ISSN 1432-0851 ; 0340-7004
    ISSN (online) 1432-0851
    ISSN 0340-7004
    DOI 10.1007/s00262-014-1650-8
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: Current Landscape and Future Directions of Biomarkers for Immunotherapy in Hepatocellular Carcinoma.

    Gok Yavuz, Betul / Hasanov, Elshad / Lee, Sunyoung S / Mohamed, Yehia I / Curran, Michael A / Koay, Eugene J / Cristini, Vittorio / Kaseb, Ahmed O

    Journal of hepatocellular carcinoma

    2021  Volume 8, Page(s) 1195–1207

    Abstract: Hepatocellular carcinoma (HCC) is the most common liver cancer and one of the leading causes of cancer-related deaths in the world. Multiple immunotherapeutic approaches have been investigated to date, and immunotherapy has become the new standard of ... ...

    Abstract Hepatocellular carcinoma (HCC) is the most common liver cancer and one of the leading causes of cancer-related deaths in the world. Multiple immunotherapeutic approaches have been investigated to date, and immunotherapy has become the new standard of care therapy in HCC. However, the current role of immunotherapy in HCC remains non-curative. Given this context, a high priority for oncology is understanding the biomarkers that predict clinical response to immunotherapy, have the potential to improve patient selection to maximize the clinical benefit, and spare unnecessary toxicity. In this review, we summarize the key predictive and prognostic biomarkers investigated in immunotherapy clinical trials in HCC and the emerging biomarkers to serve as a roadmap for future clinical trials. Biomarkers from tumoral tissues including PDL-1 expression, tissue infiltrating lymphocytes, tumor mutational burden (TMB) and specific immune signatures, and from peripheral blood including neutrophil-to-lymphocytes ratio, platelet-to-lymphocytes ratio, circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and specific cytokines, along with gut microbiota are among the studied biomarkers to date in the HCC era. More integrative approaches, including mathematical biomarkers to predict immunotherapy outcomes, are yet to be studied in HCC.
    Language English
    Publishing date 2021-09-24
    Publishing country New Zealand
    Document type Journal Article ; Review
    ZDB-ID 2780784-8
    ISSN 2253-5969
    ISSN 2253-5969
    DOI 10.2147/JHC.S322289
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top