LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 75

Search options

  1. Article: Targeting carcinoembryonic antigen-expressing tumors using a novel transcriptional and translational dual-regulated oncolytic herpes simplex virus type 1.

    Chouljenko, Dmitry V / Murad, Yanal M / Lee, I-Fang / Delwar, Zahid / Ding, Jun / Liu, Guoyu / Liu, Xiaohu / Bu, Xuexian / Sun, Yi / Samudio, Ismael / Jia, William Wei-Guo

    Molecular therapy oncolytics

    2023  Volume 28, Page(s) 334–348

    Abstract: VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing ... ...

    Abstract VG2025 is a recombinant oncolytic herpes simplex virus type 1 (HSV-1) that uses transcriptional and translational dual regulation (TTDR) of critical viral genes to enhance virus safety and promote tumor-specific virus replication without reducing virulence. The TTDR platform is based on transcriptional control of the essential HSV-1 immediate-early protein ICP27 using a tumor-specific carcinoembryonic antigen (CEA) promoter, coupled with translational control of the neurovirulence factor ICP34.5 using multiple microRNA (miR)-binding sites. VG2025 further incorporates IL-12 and the IL-15/IL-15 receptor alpha subunit complex to enhance the antitumor and immune stimulatory properties of oncolytic HSVs. The TTDR strategy was verified
    Language English
    Publishing date 2023-02-11
    Publishing country United States
    Document type Journal Article
    ISSN 2372-7705
    ISSN 2372-7705
    DOI 10.1016/j.omto.2023.02.003
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Pre-Existing HSV-1 Immunity Enhances Anticancer Efficacy of a Novel Immune-Stimulating Oncolytic Virus.

    Ding, Jun / Murad, Yanal M / Sun, Yi / Lee, I-Fang / Samudio, Ismael / Liu, Xiaohu / Jia, William Wei-Guo / Zhao, Ronghua

    Viruses

    2022  Volume 14, Issue 11

    Abstract: Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 ...

    Abstract Oncolytic viruses (OVs) can specifically replicate in the host and cause cancer cell lysis while inducing an antitumor immune response. The aim of this study is to investigate the impact of either pre-existing immunity against herpes simplex virus type-1 (HSV-1) or multicycle treatment with OVs on anticancer efficacy of VG161, an HSV-1 OV in phase 2 clinical trial. VG161 efficacy was tested in CT26 mouse models by comparing the efficacy and immune response in naïve mice or in mice that were immunized with VG161. Moreover, VG161 efficacy in HLA-matched CD34+ humanized intrahepatic cholangiocarcinoma (ICC) patient-derived xenograft (PDX) models was also tested in multicycle treatment and was compared to standard chemotherapy for this type of cancer (gemcitabine). The HSV-1-immunized mice significantly inhibited tumor growth in VG161-treated mice compared to control naïve treated mice. RNA expression profiling and ELISPOT analyses indicated changes in the tumor's immune profile in the immunized and treated group compared to naïve and treated mice, as well as enhanced T cell function depicted by higher numbers of tumor specific lymphocytes, which was enhanced by immunization. In the ICC PDX model, repeated treatment of VG161 with 2 or 3 cycles seemed to increase the anticancer efficacy of VG161. In conclusion, the anticancer efficacy of VG161 can be enhanced by pre-immunization with HSV-1 and multicycle administration when the virus is given intratumorally, indicating that pre-existing antiviral immunity might enhance OV-induced antitumor immunity. Our results suggest potential clinical benefits of HSV-1-based OV therapy in HSV-1-seropositive patients and multicycle administration of VG161 for long-term maintenance treatment.
    MeSH term(s) Humans ; Mice ; Animals ; Oncolytic Viruses/physiology ; Herpesvirus 1, Human/genetics ; Neoplasms/therapy ; Immunity ; Disease Models, Animal ; Oncolytic Virotherapy/methods
    Language English
    Publishing date 2022-10-23
    Publishing country Switzerland
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2516098-9
    ISSN 1999-4915 ; 1999-4915
    ISSN (online) 1999-4915
    ISSN 1999-4915
    DOI 10.3390/v14112327
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Targeting leukemia's "fatty tooth".

    Samudio, Ismael / Konopleva, Marina

    Blood

    2015  Volume 126, Issue 16, Page(s) 1874–1875

    Abstract: In this issue of Blood, Ricciardi et al report a novel fatty acid oxidation (FAO) inhibitor, ST1326, that effectively inhibits proliferation, survival, and chemoresistance in leukemia cell lines and primary samples. ...

    Abstract In this issue of Blood, Ricciardi et al report a novel fatty acid oxidation (FAO) inhibitor, ST1326, that effectively inhibits proliferation, survival, and chemoresistance in leukemia cell lines and primary samples.
    MeSH term(s) Carnitine/analogs & derivatives ; Carnitine O-Palmitoyltransferase/antagonists & inhibitors ; Drug Delivery Systems ; Fatty Acids/metabolism ; Humans ; Leukemia, Myeloid, Acute/drug therapy ; Neoplasm Proteins/antagonists & inhibitors
    Chemical Substances Fatty Acids ; Neoplasm Proteins ; Carnitine O-Palmitoyltransferase (EC 2.3.1.21) ; Carnitine (S7UI8SM58A)
    Language English
    Publishing date 2015-10-14
    Publishing country United States
    Document type Journal Article ; Comment
    ZDB-ID 80069-7
    ISSN 1528-0020 ; 0006-4971
    ISSN (online) 1528-0020
    ISSN 0006-4971
    DOI 10.1182/blood-2015-08-665125
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Cell-surface proteomics for the identification of novel therapeutic targets in cancer.

    Kuhlmann, Laura / Cummins, Emma / Samudio, Ismael / Kislinger, Thomas

    Expert review of proteomics

    2018  Volume 15, Issue 3, Page(s) 259–275

    Abstract: Introduction: Cancer is the second most common cause of death worldwide and its heterogeneity complicates therapy. Standard cytotoxic regiments disrupt rapidly dividing cells, regardless of their neoplastic status. The introduction of less toxic ... ...

    Abstract Introduction: Cancer is the second most common cause of death worldwide and its heterogeneity complicates therapy. Standard cytotoxic regiments disrupt rapidly dividing cells, regardless of their neoplastic status. The introduction of less toxic targeted therapies has partially contributed to the observed decrease in cancer-related mortality. Cell-surface proteins represent attractive targets for therapy, due to their easily-accessible localization and their involvement in essential signaling pathways, often dysregulated in cancer. Despite their clinical appeal, cell-surface proteins are often underrepresented in standard proteomic data sets, due to their poor solubility and lower expression levels compared to intracellular proteins. Areas covered: This review will summarize some of the available techniques for enriching the cell-surface proteome, and discuss their advantages, limitations and applicability to clinical sample-testing. Moreover, we discuss currently available strategies for the development of novel targeted therapies in cancer. Expert commentary: The interest in elucidating the cancer-associated surfaceome is growing and will likely benefit from recent advancements in instrument sensitivity, method development, and a growing body of high-quality proteomics databases. Multiomics studies, in combination with functional validations (e.g. dropout screens), and evaluation of the healthy surfaceome, will likely aid in the selection of relevant targets for future therapy development.
    MeSH term(s) Animals ; Antigens, Neoplasm/chemistry ; Antigens, Neoplasm/metabolism ; Biomarkers, Tumor/chemistry ; Biomarkers, Tumor/metabolism ; Humans ; Mass Spectrometry/methods ; Molecular Targeted Therapy/methods ; Proteomics/methods
    Chemical Substances Antigens, Neoplasm ; Biomarkers, Tumor
    Language English
    Publishing date 2018-01-23
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ZDB-ID 2299100-1
    ISSN 1744-8387 ; 1478-9450
    ISSN (online) 1744-8387
    ISSN 1478-9450
    DOI 10.1080/14789450.2018.1429924
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article ; Online: Oncolytic virotherapy in hepato-bilio-pancreatic cancer: The key to breaking the log jam?

    Li, Yuwei / Shen, Yinan / Zhao, Ronghua / Samudio, Ismael / Jia, William / Bai, Xueli / Liang, Tingbo

    Cancer medicine

    2020  Volume 9, Issue 9, Page(s) 2943–2959

    Abstract: Traditional therapies have limited efficacy in hepatocellular carcinoma, pancreatic cancer, and biliary tract cancer, especially for advanced and refractory cancers. Through a deeper understanding of antitumor immunity and the tumor microenvironment, ... ...

    Abstract Traditional therapies have limited efficacy in hepatocellular carcinoma, pancreatic cancer, and biliary tract cancer, especially for advanced and refractory cancers. Through a deeper understanding of antitumor immunity and the tumor microenvironment, novel immunotherapies are becoming available for cancer treatment. Oncolytic virus (OV) therapy is an emerging type of immunotherapy that has demonstrated effective antitumor efficacy in many preclinical studies and clinical studies. Thus, it may represent a potential feasible treatment for hard to treat gastrointestinal (GI) tumors. Here, we summarize the research progress of OV therapy for the treatment of hepato-bilio-pancreatic cancers. In general, most OV therapies exhibits potent, specific oncolysis both in cell lines in vitro and the animal models in vivo. Currently, several clinical trials have suggested that OV therapy may also be effective in patients with refractory hepato-bilio-pancreatic cancer. Multiple strategies such as introducing immunostimulatory genes, modifying virus capsid and combining various other therapeutic modalities have been shown enhanced specific oncolysis and synergistic anti-cancer immune stimulation. Combining OV with other antitumor therapies may become a more effective strategy than using virus alone. Nevertheless, more studies are needed to better understand the mechanisms underlying the therapeutic effects of OV, and to design appropriate dosing and combination strategies.
    MeSH term(s) Animals ; Biliary Tract Neoplasms/pathology ; Biliary Tract Neoplasms/therapy ; Humans ; Liver Neoplasms/pathology ; Liver Neoplasms/therapy ; Oncolytic Virotherapy/methods ; Pancreatic Neoplasms/pathology ; Pancreatic Neoplasms/therapy
    Language English
    Publishing date 2020-03-04
    Publishing country United States
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Review
    ISSN 2045-7634
    ISSN (online) 2045-7634
    DOI 10.1002/cam4.2949
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Human Fc gamma receptor IIIA blockade inhibits platelet destruction in a humanized murine model of ITP.

    Gil Gonzalez, Lazaro / Won, Kevin D / Tawhidi, Zoya / Cummins, Emma / Cruz-Leal, Yoelys / Tundidor Cabado, Yaima / Sachs, Ulrich J / Norris, Peter A A / Shan, Yuexin / Bhakta, Varsha / Li, Janessa / Samudio, Ismael / Silva-Moreno, Begonia / Cerna-Portillo, Liza / Pavon Oro, Alequis / Bergqvist, Peter / Chan, Patrick / Moorehead, Amy / Sholzberg, Michelle /
    Sheffield, William P / Lazarus, Alan H

    Blood advances

    2024  Volume 8, Issue 8, Page(s) 1869–1879

    Abstract: Abstract: Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on ... ...

    Abstract Abstract: Fc gamma receptor (FcγR) IIIA is an important receptor for immunoglobulin G (IgG) and is involved in immune defense mechanisms as well as tissue destruction in some autoimmune diseases including immune thrombocytopenia (ITP). FcγRIIIA on macrophages can trigger phagocytosis of IgG-sensitized platelets, and prior pilot studies observed blockade of FcγRIIIA increased platelet counts in patients with ITP. Unfortunately, although blockade of FcγRIIIA in patients with ITP increased platelet counts, its engagement by the blocking antibody drove serious adverse inflammatory reactions. These adverse events were postulated to originate from the antibody's Fc and/or bivalent nature. The blockade of human FcγRIIIA in vivo with a monovalent construct lacking an active Fc region has not yet been achieved. To effectively block FcγRIIIA in vivo, we developed a high affinity monovalent single-chain variable fragment (scFv) that can bind and block human FcγRIIIA. This scFv (17C02) was expressed in 3 formats: a monovalent fusion protein with albumin, a 1-armed human IgG1 antibody, and a standard bivalent mouse (IgG2a) antibody. Both monovalent formats were effective in preventing phagocytosis of ITP serum-sensitized human platelets. In vivo studies using FcγR-humanized mice demonstrated that both monovalent therapeutics were also able to increase platelet counts. The monovalent albumin fusion protein did not have adverse event activity as assessed by changes in body temperature, whereas the 1-armed antibody induced some changes in body temperature even though the Fc region function was impaired by the Leu234Ala and Leu235Ala mutations. These data demonstrate that monovalent blockade of human FcγRIIIA in vivo can potentially be a therapeutic strategy for patients with ITP.
    MeSH term(s) Humans ; Mice ; Animals ; Purpura, Thrombocytopenic, Idiopathic/drug therapy ; Receptors, IgG/metabolism ; Disease Models, Animal ; Thrombocytopenia ; Immunoglobulin G/therapeutic use ; Albumins/therapeutic use
    Chemical Substances Receptors, IgG ; Immunoglobulin G ; Albumins
    Language English
    Publishing date 2024-02-06
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2915908-8
    ISSN 2473-9537 ; 2473-9529
    ISSN (online) 2473-9537
    ISSN 2473-9529
    DOI 10.1182/bloodadvances.2023012155
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Asparaginase unveils glutamine-addicted AML.

    Samudio, Ismael / Konopleva, Marina

    Blood

    2013  Volume 122, Issue 20, Page(s) 3398–3400

    Abstract: In this issue of Blood, Willems et al describe the dependence of acute myeloid leukemia (AML) cells on glutamine for maintaining protein synthesis downstream of mammalian target of rapamycin (mTOR) and show that the enzyme asparaginase can be used to ... ...

    Abstract In this issue of Blood, Willems et al describe the dependence of acute myeloid leukemia (AML) cells on glutamine for maintaining protein synthesis downstream of mammalian target of rapamycin (mTOR) and show that the enzyme asparaginase can be used to target this dependence. Using various AML cell lines, primary samples, and CD341 stem cells from healthy donors, the authors support the notion that asparaginase may offer a therapeutic benefit in AML—not from its well-known enzymatic activity, but from its “off-target” effects on glutamine levels that result in inhibition of downstream mTOR signaling, inhibition of protein synthesis, and ultimately loss of viability.
    MeSH term(s) Animals ; Female ; Glutamine/antagonists & inhibitors ; Humans ; Leukemia, Myeloid, Acute/drug therapy ; Male
    Chemical Substances Glutamine (0RH81L854J)
    Language English
    Publishing date 2013-11-13
    Publishing country United States
    Document type Journal Article ; Comment
    ZDB-ID 80069-7
    ISSN 1528-0020 ; 0006-4971
    ISSN (online) 1528-0020
    ISSN 0006-4971
    DOI 10.1182/blood-2013-09-526392
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article: Targeting a Tumor-Specific Epitope on Podocalyxin Increases Survival in Human Tumor Preclinical Models.

    Canals Hernaez, Diana / Hughes, Michael R / Li, Yicong / Mainero Rocca, Ilaria / Dean, Pamela / Brassard, Julyanne / Bell, Erin M / Samudio, Ismael / Mes-Masson, Anne-Marie / Narimatsu, Yoshiki / Clausen, Henrik / Blixt, Ola / Roskelley, Calvin D / McNagny, Kelly M

    Frontiers in oncology

    2022  Volume 12, Page(s) 856424

    Abstract: Podocalyxin (Podxl) is a CD34-related cell surface sialomucin that is normally highly expressed by adult vascular endothelia and kidney podocytes where it plays a key role in blocking adhesion. Importantly, it is also frequently upregulated on a wide ... ...

    Abstract Podocalyxin (Podxl) is a CD34-related cell surface sialomucin that is normally highly expressed by adult vascular endothelia and kidney podocytes where it plays a key role in blocking adhesion. Importantly, it is also frequently upregulated on a wide array of human tumors and its expression often correlates with poor prognosis. We previously showed that, in xenograft studies, Podxl plays a key role in metastatic disease by making tumor initiating cells more mobile and invasive. Recently, we developed a novel antibody, PODO447, which shows exquisite specificity for a tumor-restricted glycoform of Podxl but does not react with Podxl expressed by normal adult tissue. Here we utilized an array of glycosylation defective cell lines to further define the PODO447 reactive epitope and reveal it as an O-linked core 1 glycan presented in the context of the Podxl peptide backbone. Further, we show that when coupled to monomethyl auristatin E (MMAE) toxic payload, PODO447 functions as a highly specific and effective antibody drug conjugate (ADC) in killing ovarian, pancreatic, glioblastoma and leukemia cell lines
    Language English
    Publishing date 2022-05-04
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2649216-7
    ISSN 2234-943X
    ISSN 2234-943X
    DOI 10.3389/fonc.2022.856424
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article ; Online: The mitochondria target drug avocatin B synergizes with induction chemotherapeutics to induce leukemia cell death.

    Tcheng, Matthew / Samudio, Ismael / Lee, Eric A / Minden, Mark D / Spagnuolo, Paul A

    Leukemia & lymphoma

    2016  Volume 58, Issue 4, Page(s) 986–988

    MeSH term(s) Antineoplastic Agents/pharmacology ; Cell Death/drug effects ; Cell Line, Tumor ; Drug Synergism ; Fatty Acids/metabolism ; Humans ; Leukemia/drug therapy ; Leukemia/metabolism ; Mitochondria/drug effects ; Mitochondria/metabolism ; Oxidation-Reduction/drug effects
    Chemical Substances Antineoplastic Agents ; Fatty Acids
    Language English
    Publishing date 2016-08-25
    Publishing country United States
    Document type Letter ; Research Support, Non-U.S. Gov't
    ZDB-ID 1042374-6
    ISSN 1029-2403 ; 1042-8194
    ISSN (online) 1029-2403
    ISSN 1042-8194
    DOI 10.1080/10428194.2016.1218005
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article: Induction of Durable Antitumor Response by a Novel Oncolytic Herpesvirus Expressing Multiple Immunomodulatory Transgenes.

    Chouljenko, Dmitry V / Ding, Jun / Lee, I-Fang / Murad, Yanal M / Bu, Xuexian / Liu, Guoyu / Delwar, Zahid / Sun, Yi / Yu, Sheng / Samudio, Ismael / Zhao, Ronghua / Jia, William Wei-Guo

    Biomedicines

    2020  Volume 8, Issue 11

    Abstract: Oncolytic virotherapy is a promising new tool for cancer treatment, but direct lytic destruction of tumor cells is not sufficient and must be accompanied by strong immune activation to elicit anti-tumor immunity. We report here the creation of a novel ... ...

    Abstract Oncolytic virotherapy is a promising new tool for cancer treatment, but direct lytic destruction of tumor cells is not sufficient and must be accompanied by strong immune activation to elicit anti-tumor immunity. We report here the creation of a novel replication-competent recombinant oncolytic herpes simplex virus type 1 (VG161) that carries genes coding for IL-12, IL-15, and IL-15 receptor alpha subunit, along with a peptide fusion protein capable of disrupting PD-1/PD-L1 interactions. The VG161 virus replicates efficiently and exhibits robust cytotoxicity in multiple tumor cell lines. Moreover, the encoded cytokines and the PD-L1 blocking peptide work cooperatively to boost immune cell function. In vivo testing in syngeneic CT26 and A20 tumor models reveals superior efficacy when compared to a backbone virus that does not express exogenous genes. Intratumoral injection of VG161 induces abscopal responses in non-injected distal tumors and grants resistance to tumor re-challenge. The robust anti-tumor effect of VG161 is associated with T cell and NK cell tumor infiltration, expression of Th1 associated genes in the injection site, and increased frequency of splenic tumor-specific T cells. VG161 also displayed a superb safety profile in GLP acute and repeated injection toxicity studies performed using cynomolgus monkeys. Overall, we demonstrate that VG161 can induce robust oncolysis and stimulate a robust anti-tumor immune response without sacrificing safety.
    Language English
    Publishing date 2020-11-09
    Publishing country Switzerland
    Document type Journal Article
    ZDB-ID 2720867-9
    ISSN 2227-9059
    ISSN 2227-9059
    DOI 10.3390/biomedicines8110484
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top