1
|
Haller SD, Essani K. Oncolytic Tanapoxvirus Variants Expressing mIL-2 and mCCL-2 Regress Human Pancreatic Cancer Xenografts in Nude Mice. Biomedicines 2024; 12:1834. [PMID: 39200298 PMCID: PMC11351728 DOI: 10.3390/biomedicines12081834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fifth leading cause of cancer-related death and presents the lowest 5-year survival rate of any form of cancer in the US. Only 20% of PDAC patients are suitable for surgical resection and adjuvant chemotherapy, which remains the only curative treatment. Chemotherapeutic and gene therapy treatments are associated with adverse effects and lack specificity/efficacy. In this study, we assess the oncolytic potential of immuno-oncolytic tanapoxvirus (TPV) recombinants expressing mouse monocyte chemoattractant protein (mMCP-1 or mCCL2) and mouse interleukin (mIL)-2 in human pancreatic BxPc-3 cells using immunocompromised and CD-3+ T-cell-reconstituted mice. Intratumoral treatment with TPV/∆66R/mCCL2 and TPV/∆66R/mIL-2 resulted in a regression in BxPc-3 xenograft volume compared to control in immunocompromised mice; mCCL-2 expressing TPV OV resulted in a significant difference from control at p < 0.05. Histological analysis of immunocompromised mice treated with TPV/∆66R/mCCL2 or TPV/∆66R/mIL-2 demonstrated multiple biomarkers indicative of increased severity of chronic, active inflammation compared to controls. In conclusion, TPV recombinants expressing mCCL2 and mIL-2 demonstrated a therapeutic effect via regression in BxPc-3 tumor xenografts. Considering the enhanced oncolytic potency of TPV recombinants demonstrated against PDAC in this study, further investigation as an alternative or combination treatment option for human PDAC may be warranted.
Collapse
Affiliation(s)
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008-5410, USA;
| |
Collapse
|
2
|
Salvato I, Marchini A. Immunotherapeutic Strategies for the Treatment of Glioblastoma: Current Challenges and Future Perspectives. Cancers (Basel) 2024; 16:1276. [PMID: 38610954 PMCID: PMC11010873 DOI: 10.3390/cancers16071276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Despite decades of research and the best up-to-date treatments, grade 4 Glioblastoma (GBM) remains uniformly fatal with a patient median overall survival of less than 2 years. Recent advances in immunotherapy have reignited interest in utilizing immunological approaches to fight cancer. However, current immunotherapies have so far not met the anticipated expectations, achieving modest results in their journey from bench to bedside for the treatment of GBM. Understanding the intrinsic features of GBM is of crucial importance for the development of effective antitumoral strategies to improve patient life expectancy and conditions. In this review, we provide a comprehensive overview of the distinctive characteristics of GBM that significantly influence current conventional therapies and immune-based approaches. Moreover, we present an overview of the immunotherapeutic strategies currently undergoing clinical evaluation for GBM treatment, with a specific emphasis on those advancing to phase 3 clinical studies. These encompass immune checkpoint inhibitors, adoptive T cell therapies, vaccination strategies (i.e., RNA-, DNA-, and peptide-based vaccines), and virus-based approaches. Finally, we explore novel innovative strategies and future prospects in the field of immunotherapy for GBM.
Collapse
Affiliation(s)
- Ilaria Salvato
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg;
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
3
|
Semenov DV, Vasileva NS, Dymova MA, Mishinov SV, Savinovskaya YI, Ageenko AB, Dome AS, Zinchenko ND, Stepanov GA, Kochneva GV, Richter VA, Kuligina EV. Transcriptome Changes in Glioma Cells upon Infection with the Oncolytic Virus VV-GMCSF-Lact. Cells 2023; 12:2616. [PMID: 37998351 PMCID: PMC10670333 DOI: 10.3390/cells12222616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/25/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
Oncolytic virotherapy is a rapidly evolving approach that aims to selectively kill cancer cells. We designed a promising recombinant vaccinia virus, VV-GMCSF-Lact, for the treatment of solid tumors, including glioma. We assessed how VV-GMCSF-Lact affects human cells using immortalized and patient-derived glioma cultures and a non-malignant brain cell culture. Studying transcriptome changes in cells 12 h or 24 h after VV-GMCSF-Lact infection, we detected the common activation of histone genes. Additionally, genes associated with the interferon-gamma response, NF-kappa B signaling pathway, and inflammation mediated by chemokine and cytokine signaling pathways showed increased expression. By contrast, genes involved in cell cycle progression, including spindle organization, sister chromatid segregation, and the G2/M checkpoint, were downregulated following virus infection. The upregulation of genes responsible for Golgi vesicles, protein transport, and secretion correlated with reduced sensitivity to the cytotoxic effect of VV-GMCSF-Lact. Higher expression of genes encoding proteins, which participate in the maturation of pol II nuclear transcripts and mRNA splicing, was associated with an increased sensitivity to viral cytotoxicity. Genes whose expression correlates with the sensitivity of cells to the virus are important for increasing the effectiveness of cancer virotherapy. Overall, the results highlight molecular markers, biological pathways, and gene networks influencing the response of glioma cells to VV-GMCSF-Lact.
Collapse
Affiliation(s)
- Dmitriy V. Semenov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Natalia S. Vasileva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Maya A. Dymova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Sergey V. Mishinov
- Novosibirsk Research Institute of Traumatology and Orthopedics n.a. Ya.L. Tsivyan, Department of Neurosurgery, Frunze Street, 17, 630091 Novosibirsk, Russia;
| | - Yulya I. Savinovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Alisa B. Ageenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Anton S. Dome
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Nikita D. Zinchenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Grigory A. Stepanov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Galina V. Kochneva
- State Research Center of Virology and Biotechnology “Vector”, Rospotrebnadzor, 630559 Koltsovo, Russia;
| | - Vladimir A. Richter
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| | - Elena V. Kuligina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentyev Avenue, 8, 630090 Novosibirsk, Russia; (N.S.V.); (M.A.D.); (Y.I.S.); (A.B.A.); (A.S.D.); (N.D.Z.); (G.A.S.); (V.A.R.); (E.V.K.)
| |
Collapse
|
4
|
Huang S, Ren L, Beck JA, Phelps TE, Olkowski C, Ton A, Roy J, White ME, Adler S, Wong K, Cherukuri A, Zhang X, Basuli F, Choyke PL, Jagoda EM, LeBlanc AK. Exploration of Imaging Biomarkers for Metabolically-Targeted Osteosarcoma Therapy in a Murine Xenograft Model. Cancer Biother Radiopharm 2023; 38:475-485. [PMID: 37253167 PMCID: PMC10623067 DOI: 10.1089/cbr.2022.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023] Open
Abstract
Background: Osteosarcoma (OS) is an aggressive pediatric cancer with unmet therapeutic needs. Glutaminase 1 (GLS1) inhibition, alone and in combination with metformin, disrupts the bioenergetic demands of tumor progression and metastasis, showing promise for clinical translation. Materials and Methods: Three positron emission tomography (PET) clinical imaging agents, [18F]fluoro-2-deoxy-2-D-glucose ([18F]FDG), 3'-[18F]fluoro-3'-deoxythymidine ([18F]FLT), and (2S, 4R)-4-[18F]fluoroglutamine ([18F]GLN), were evaluated in the MG63.3 human OS xenograft mouse model, as companion imaging biomarkers after treatment for 7 d with a selective GLS1 inhibitor (CB-839, telaglenastat) and metformin, alone and in combination. Imaging and biodistribution data were collected from tumors and reference tissues before and after treatment. Results: Drug treatment altered tumor uptake of all three PET agents. Relative [18F]FDG uptake decreased significantly after telaglenastat treatment, but not within control and metformin-only groups. [18F]FLT tumor uptake appears to be negatively affected by tumor size. Evidence of a flare effect was seen with [18F]FLT imaging after treatment. Telaglenastat had a broad influence on [18F]GLN uptake in tumor and normal tissues. Conclusions: Image-based tumor volume quantification is recommended for this paratibial tumor model. The performance of [18F]FLT and [18F]GLN was affected by tumor size. [18F]FDG may be useful in detecting telaglenastat's impact on glycolysis. Exploration of kinetic tracer uptake protocols is needed to define clinically relevant patterns of [18F]GLN uptake in patients receiving telaglenastat.
Collapse
Affiliation(s)
- Shan Huang
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ling Ren
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jessica A. Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tim E. Phelps
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Colleen Olkowski
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anita Ton
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jyoti Roy
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Margaret E. White
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Bethesda, Maryland, USA
| | - Karen Wong
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Aswini Cherukuri
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Xiang Zhang
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Elaine M. Jagoda
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amy K. LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Riederer S, Del Canizo A, Navas J, Peter MG, Link EK, Sutter G, Rojas JJ. Improving poxvirus-mediated antitumor immune responses by deleting viral cGAMP-specific nuclease. Cancer Gene Ther 2023:10.1038/s41417-023-00610-5. [PMID: 37016144 DOI: 10.1038/s41417-023-00610-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/21/2023] [Accepted: 03/21/2023] [Indexed: 04/06/2023]
Abstract
cGAMP-specific nucleases (poxins) are a recently described family of proteins dedicated to obstructing cyclic GMP-AMP synthase signaling (cGAS), an important sensor triggered by cytoplasmic viral replication that activates type I interferon (IFN) production. The B2R gene of vaccinia viruses (VACV) codes for one of these nucleases. Here, we evaluated the effects of inactivating the VACV B2 nuclease in the context of an oncolytic VACV. VACV are widely used as anti-cancer vectors due to their capacity to activate immune responses directed against tumor antigens. We aimed to elicit robust antitumor immunity by preventing viral inactivation of the cGAS/STING/IRF3 pathway after infection of cancer cells. Activation of such a pathway is associated with a dominant T helper 1 (Th1) cell differentiation of the response, which benefits antitumor outcomes. Deletion of the B2R gene resulted in enhanced IRF3 phosphorylation and type I IFN expression after infection of tumor cells, while effective VACV replication remained unimpaired, both in vitro and in vivo. In syngeneic mouse tumor models, the absence of the VACV cGAMP-specific nuclease translated into improved antitumor activity, which was associated with antitumor immunity directed against tumor epitopes.
Collapse
Affiliation(s)
- Stephanie Riederer
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Ana Del Canizo
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona-UB, Barcelona, Spain
- Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Navas
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona-UB, Barcelona, Spain
- Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Marlowe G Peter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Ellen K Link
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany
| | - Gerd Sutter
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany.
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| | - Juan J Rojas
- Division of Virology, Department of Veterinary Sciences, LMU Munich, Munich, Germany.
- Immunology Unit, Department of Pathology and Experimental Therapies, School of Medicine, University of Barcelona-UB, Barcelona, Spain.
- Immunity, Inflammation, and Cancer Group, Oncobell Program, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
6
|
Shakiba Y, Vorobyev PO, Naumenko VA, Kochetkov DV, Zajtseva KV, Valikhov MP, Yusubalieva GM, Gumennaya YD, Emelyanov EA, Semkina AS, Baklaushev VP, Chumakov PM, Lipatova AV. Oncolytic Efficacy of a Recombinant Vaccinia Virus Strain Expressing Bacterial Flagellin in Solid Tumor Models. Viruses 2023; 15:828. [PMID: 37112810 PMCID: PMC10142208 DOI: 10.3390/v15040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
Oncolytic viral therapy is a promising novel approach to cancer treatment. Oncolytic viruses cause tumor regression through direct cytolysis on the one hand and recruiting and activating immune cells on the other. In this study, to enhance the antitumor efficacy of the thymidine kinase-deficient vaccinia virus (VV, Lister strain), recombinant variants encoding bacterial flagellin (subunit B) of Vibrio vulnificus (LIVP-FlaB-RFP), firefly luciferase (LIVP-Fluc-RFP) or red fluorescent protein (LIVP-RFP) were developed. The LIVP-FLuc-RFP strain demonstrated exceptional onco-specificity in tumor-bearing mice, detected by the in vivo imaging system (IVIS). The antitumor efficacy of these variants was explored in syngeneic murine tumor models (B16 melanoma, CT26 colon cancer and 4T1 breast cancer). After intravenous treatment with LIVP-FlaB-RFP or LIVP-RFP, all mice tumor models exhibited tumor regression, with a prolonged survival rate in comparison with the control mice. However, superior oncolytic activity was observed in the B16 melanoma models treated with LIVP-FlaB-RFP. Tumor-infiltrated lymphocytes and the cytokine analysis of the serum and tumor samples from the melanoma-xenografted mice treated with these virus variants demonstrated activation of the host's immune response. Thus, the expression of bacterial flagellin by VV can enhance its oncolytic efficacy against immunosuppressive solid tumors.
Collapse
Affiliation(s)
- Yasmin Shakiba
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia;
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Pavel O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Victor A. Naumenko
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
| | - Dmitry V. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ksenia V. Zajtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marat P. Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Yana D. Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Egor A. Emelyanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
7
|
Abstract
The cell cycle is the series of events that take place in a cell that drives it to divide and produce two new daughter cells. Through more than 100 years of efforts by scientists, we now have a much clearer picture of cell cycle progression and its regulation. The typical cell cycle in eukaryotes is composed of the G1, S, G2, and M phases. The M phase is further divided into prophase, prometaphase, metaphase, anaphase, telophase, and cytokinesis. Cell cycle progression is mediated by cyclin-dependent kinases (Cdks) and their regulatory cyclin subunits. However, the driving force of cell cycle progression is growth factor-initiated signaling pathways that controls the activity of various Cdk-cyclin complexes. Most cellular events, including DNA duplication, gene transcription, protein translation, and post-translational modification of proteins, occur in a cell-cycle-dependent manner. To understand these cellular events and their underlying molecular mechanisms, it is desirable to have a population of cells that are traversing the cell cycle synchronously. This can be achieved through a process called cell synchronization. Many methods have been developed to synchronize cells to the various phases of the cell cycle. These methods could be classified into two groups: synchronization methods using chemical inhibitors and synchronization methods without using chemical inhibitors. All these methods have their own merits and shortcomings.
Collapse
Affiliation(s)
- Zhixiang Wang
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
8
|
|
9
|
Oncolytic virotherapy: Challenges and solutions. Curr Probl Cancer 2021; 45:100639. [DOI: 10.1016/j.currproblcancer.2020.100639] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 07/22/2020] [Indexed: 12/16/2022]
|
10
|
Gallardo F, Schmitt D, Brandely R, Brua C, Silvestre N, Findeli A, Foloppe J, Top S, Kappler-Gratias S, Quentin-Froignant C, Morin R, Lagarde JM, Bystricky K, Bertagnoli S, Erbs P. Fluorescent Tagged Vaccinia Virus Genome Allows Rapid and Efficient Measurement of Oncolytic Potential and Discovery of Oncolytic Modulators. Biomedicines 2020; 8:biomedicines8120543. [PMID: 33256205 PMCID: PMC7760631 DOI: 10.3390/biomedicines8120543] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022] Open
Abstract
As a live biologic agent, oncolytic vaccinia virus has the ability to target and selectively amplify at tumor sites. We have previously reported that deletion of thymidine kinase and ribonucleotide reductase genes in vaccinia virus can increase the safety and efficacy of the virus. Here, to allow direct visualization of the viral genome in living cells, we incorporated the ANCH target sequence and the OR3-Santaka gene in the double-deleted vaccinia virus. Infection of human tumor cells with ANCHOR3-tagged vaccinia virus enables visualization and quantification of viral genome dynamics in living cells. The results show that the ANCHOR technology permits the measurement of the oncolytic potential of the double deleted vaccinia virus. Quantitative analysis of infection kinetics and of viral DNA replication allow rapid and efficient identification of inhibitors and activators of oncolytic activity. Our results highlight the potential application of the ANCHOR technology to track vaccinia virus and virtually any kind of poxvirus in living cells.
Collapse
Affiliation(s)
- Franck Gallardo
- NeoVirTech SAS, 31106 Toulouse, France; (S.T.); (S.K.-G.); (C.Q.-F.)
- Correspondence: (F.G.); (P.E.)
| | - Doris Schmitt
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Renée Brandely
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Catherine Brua
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Nathalie Silvestre
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Annie Findeli
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Johann Foloppe
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
| | - Sokunthea Top
- NeoVirTech SAS, 31106 Toulouse, France; (S.T.); (S.K.-G.); (C.Q.-F.)
| | | | | | - Renaud Morin
- Imactiv-3D SAS, 31106 Toulouse, France; (R.M.); (J.-M.L.)
| | | | - Kerstin Bystricky
- Centre de Biologie Intégrative (CBI), Laboratoire de Biologie Moléculaire Eucaryote (LBME), University of Toulouse, UPS, CNRS, 31400 Toulouse, France;
- Institut Universitaire de France (IUF), 75231 Paris, France
| | | | - Philippe Erbs
- Transgene SA, 67405 Illkirch-Graffenstaden, France; (D.S.); (R.B.); (C.B.); (N.S.); (A.F.); (J.F.)
- Correspondence: (F.G.); (P.E.)
| |
Collapse
|
11
|
Hammad M, Cornejo YR, Batalla-Covello J, Majid AA, Burke C, Liu Z, Yuan YC, Li M, Dellinger TH, Lu J, Chen NG, Fong Y, Aboody KS, Mooney R. Neural Stem Cells Improve the Delivery of Oncolytic Chimeric Orthopoxvirus in a Metastatic Ovarian Cancer Model. Mol Ther Oncolytics 2020; 18:326-334. [PMID: 32775617 PMCID: PMC7394740 DOI: 10.1016/j.omto.2020.07.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/01/2020] [Indexed: 12/22/2022] Open
Abstract
Oncolytic virotherapy represents a promising approach for treating recurrent and/or drug-resistant ovarian cancer. However, its successful application in the clinic has been hampered by rapid immune-mediated clearance, which reduces viral delivery to the tumor. Patient-derived mesenchymal stem cells that home to tumors have been used as viral delivery tools, but variability associated with autologous cell isolations limits the clinical applicability of this approach. We previously developed an allogeneic, clonal neural stem cell (NSC) line (HB1.F3.CD21) that can be used to deliver viral cargo. Here, we demonstrate that this NSC line can improve the delivery of a thymidine kinase gene-deficient conditionally replication-competent orthopoxvirus, CF33, in a preclinical cisplatin-resistant peritoneal ovarian metastases model. Overall, our findings provide the basis for using off-the-shelf allogeneic cell-based delivery platforms for oncolytic viruses, thus providing a more efficient delivery alternative compared with the free virus administration approach.
Collapse
Affiliation(s)
- Mohamed Hammad
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
| | - Yvonne R. Cornejo
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
- Irell & Manella Graduate School for Biological Sciences at the Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Jennifer Batalla-Covello
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
- Irell & Manella Graduate School for Biological Sciences at the Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Asma Abdul Majid
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
| | - Connor Burke
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
| | - Zheng Liu
- Translational Bioinformatics Division, Center for Informatics, City of Hope, Duarte, CA 91010, USA
| | - Yate-Ching Yuan
- Translational Bioinformatics Division, Center for Informatics, City of Hope, Duarte, CA 91010, USA
| | - Min Li
- Department of Information Sciences, Division of Biostatistics at the Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Thanh H. Dellinger
- Division of Gynecologic Surgery, Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Jianming Lu
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
| | - Nanhai G. Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
- Center for Gene Therapy, City of Hope, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope, Duarte, CA 91010, USA
- Center for Gene Therapy, City of Hope, Duarte, CA 91010, USA
| | - Karen S. Aboody
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
- Division of Neurosurgery, City of Hope, Duarte, CA 91010, USA
| | - Rachael Mooney
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
12
|
Li Y, Shen Y, Zhao R, Samudio I, Jia W, Bai X, Liang T. Oncolytic virotherapy in hepato-bilio-pancreatic cancer: The key to breaking the log jam? Cancer Med 2020; 9:2943-2959. [PMID: 32130786 PMCID: PMC7196045 DOI: 10.1002/cam4.2949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
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.
Collapse
Affiliation(s)
- Yuwei Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.,Innovation Center for the study of Pancreatic Diseases, Hangzhou, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.,Innovation Center for the study of Pancreatic Diseases, Hangzhou, China
| | | | | | - William Jia
- Virogin Biotech Canada Ltd, Vancouver, Canada
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.,Innovation Center for the study of Pancreatic Diseases, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China.,Innovation Center for the study of Pancreatic Diseases, Hangzhou, China
| |
Collapse
|
13
|
Foloppe J, Kempf J, Futin N, Kintz J, Cordier P, Pichon C, Findeli A, Vorburger F, Quemeneur E, Erbs P. The Enhanced Tumor Specificity of TG6002, an Armed Oncolytic Vaccinia Virus Deleted in Two Genes Involved in Nucleotide Metabolism. MOLECULAR THERAPY-ONCOLYTICS 2019; 14:1-14. [PMID: 31011628 PMCID: PMC6461584 DOI: 10.1016/j.omto.2019.03.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 03/18/2019] [Indexed: 11/25/2022]
Abstract
Oncolytic vaccinia viruses are currently in clinical development. However, the safety and the tumor selectivity of these oncolytic viruses must be improved. We previously constructed a first-generation oncolytic vaccinia virus by expressing the suicide gene FCU1 inserted in the J2R locus that encodes thymidine kinase. We demonstrated that the combination of this thymidine-kinase-deleted vaccinia virus and the FCU1/5-fluocytosine system is a potent vector for cancer therapy. Here, we developed a second generation of vaccinia virus, named TG6002, expressing FCU1 and with targeted deletions of the J2R gene and the I4L gene, which encodes the large subunit of the ribonucleotide reductase. Compared to the previously used single thymidine-kinase-deleted vaccinia virus, TG6002 is highly attenuated in normal cells, yet it displays tumor-selective replication and tumor cell killing. TG6002 replication is highly dependent on cellular ribonucleotide reductase levels and is less pathogenic than the single-deleted vaccinia virus. Tumor-selective viral replication, prolonged therapeutic levels of 5-fluorouracil in tumors, and significant antitumor effects were observed in multiple human xenograft tumor models after systemic injection of TG6002 and 5-fluorocytosine. TG6002 displays a convincing safety profile and is a promising candidate for treatment of cancer in humans.
Collapse
|
14
|
Yang X, Huang B, Deng L, Hu Z. Progress in gene therapy using oncolytic vaccinia virus as vectors. J Cancer Res Clin Oncol 2018; 144:2433-2440. [PMID: 30293118 DOI: 10.1007/s00432-018-2762-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/28/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Vaccinia virus was widely used in the World Health Organization's smallpox eradication campaign and is currently a promising vector for gene therapy owing to its unique characteristics. Vaccinia virus can selectively replicate and propagate productively in tumor cells, resulting in oncolysis. In addition, rapid viral particle production, wide host range, large genome size (approximately 200 kb), and safe handling render vaccinia virus a suitable vector for gene therapy. MATERIALS AND METHODS Cancer vaccines and gene therapy are being studied in clinical trials and experiment researches. However, we put forward unique challenges of optimal selection of foreign genes, administration and modification of VACV, personalized medicine, and other existing problems, based on current researches and our own experiments. CONCLUSION This review presents an overview of the vaccinia virus from its mechanisms to medical researches and clinical trials. We believe that the solution to these problems will contribute to understanding mechanisms of VACV and provide a theoretical basis for clinical treatment.
Collapse
Affiliation(s)
- Xue Yang
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China
| | - Biao Huang
- School of Life Science, Zhejiang Sci-Tech University, Zhejiang, 310018, China
| | - Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China
| | - Zhigang Hu
- Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, 214023, Jiangsu, China.
| |
Collapse
|
15
|
Ricordel M, Foloppe J, Pichon C, Findeli A, Tosch C, Cordier P, Cochin S, Quémeneur E, Camus-Bouclainville C, Bertagnoli S, Erbs P. Oncolytic properties of non-vaccinia poxviruses. Oncotarget 2018; 9:35891-35906. [PMID: 30542506 PMCID: PMC6267605 DOI: 10.18632/oncotarget.26288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/24/2018] [Indexed: 01/13/2023] Open
Abstract
Vaccinia virus, a member of the Poxviridae family, has been extensively used as an oncolytic agent and has entered late stage clinical development. In this study, we evaluated the potential oncolytic properties of other members of the Poxviridae family. Numerous tumor cell lines were infected with ten non-vaccinia poxviruses to identify which virus displayed the most potential as an oncolytic agent. Cell viability indicated that tumor cell lines were differentially susceptible to each virus. Raccoonpox virus was the most potent of the tested poxviruses and was highly effective in controlling cell growth in all tumor cell lines. To investigate further the oncolytic capacity of the Raccoonpox virus, we have generated a thymidine kinase (TK)-deleted recombinant Raccoonpox virus expressing the suicide gene FCU1. This TK-deleted Raccoonpox virus was notably attenuated in normal primary cells but replicated efficiently in numerous tumor cell lines. In human colon cancer xenograft model, a single intratumoral inoculation of the recombinant Raccoonpox virus, in combination with 5-fluorocytosine administration, produced relevant tumor growth control. The results demonstrated significant antitumoral activity of this new modified Raccoonpox virus armed with FCU1 and this virus could be considered to be included into the growing armamentarium of oncolytic virotherapy for cancer.
Collapse
Affiliation(s)
- Marine Ricordel
- Transgene SA, Illkirch-Graffenstaden 67405, France.,Current address: Polyplus-transfection SA, Illkirch-Graffenstaden 67400, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Deng L, Fan J, Ding Y, Zhang J, Zhou B, Zhang Y, Huang B. Oncolytic efficacy of thymidine kinase-deleted vaccinia virus strain Guang9. Oncotarget 2018; 8:40533-40543. [PMID: 28465492 PMCID: PMC5522336 DOI: 10.18632/oncotarget.17125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 11/29/2022] Open
Abstract
Oncolytic virotherapy is being developed as a promising platform for cancer therapy due to its ability to lyse cancer cells in a tumor-specific manner. Vaccinia virus has been used as a live vaccine in the smallpox eradication program and now is being potential in cancer therapy with a great safety profile. Vaccinia strain Guang9 (VG9) is an attenuated Chinese vaccinia virus and its oncolytic efficacy has been evaluated in our previous study. To improve the tumor selectivity and oncolytic efficacy, we here developed a thymidine kinase (TK)-deleted vaccinia virus based on Guang9 strain. The viral replication, marker gene expression and cytotoxicity in various cell lines were evaluated; antitumor effects in vivo were assessed in multiple tumor models. In vitro, the TK-deleted vaccinia virus replicated rapidly, but the cytotoxicity varied in different cell lines. It was notably attenuated in normal cells and resting cells in vitro, while tumor-selectively replicated in vivo. Significant antitumor effects were observed both in murine melanoma tumor model and human hepatoma tumor model. It significantly inhibited the growth of subcutaneously implanted tumors and prolonged the survival of tumor-bearing mice. Collectively, TK-deleted vaccinia strain Guang9 is a promising constructive virus vector for tumor-directed gene therapy and will be a potential therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jun Fan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yuedi Ding
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jue Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Bin Zhou
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yi Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Biao Huang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| |
Collapse
|
17
|
He Q, Zou L, Zhang PA, Lui JX, Skog S, Fornander T. The Clinical Significance of Thymidine Kinase 1 Measurement in Serum of Breast Cancer Patients Using Anti-TK1 Antibody. Int J Biol Markers 2018; 15:139-46. [PMID: 10883887 DOI: 10.1177/172460080001500203] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The activity of total thymidine kinase in serum (S-TK) has been used as a tumor maker for decades. To date such activity has been determined using [125]I-iodo-deoxyuridine as a substrate. The aim of this study was to develop a new, antibody-based technique for the measurement of cytoplasmic thymidine kinase (TK1) in serum. Both mono- and polyclonal antibodies against S-TK1 were used in dot blot assay. S-TK1 was characterized by SDS and IEF techniques. Sixty-five breast cancer patients were studied, including 17 preoperative and 38 postoperative tumor-free patients and 10 patients with metastases to the lymph nodes (N1–2). They were compared to patients with benign tumors (n=21) and healthy volunteers (n=11). S-TK1 was low (0–1.0 pM) in healthy volunteers, while in preoperative patients the level was increased 6–110-fold. Significant differences were observed between preoperative patients and healthy volunteers (p=0.005), preoperative patients and patients with benign tumors (p<0.001), and preoperative patients and postoperative patients without metastases (p<0.001). No significant difference was observed between preoperative patients and postoperative patients with metastases (p=0.191). The S-TK activity in preoperative patients was also high in serum, but no decrease was observed following surgery. In conclusion, the anti-TK1 antibody could be a good marker for monitoring the response of breast cancer patients to therapy.
Collapse
Affiliation(s)
- Q He
- Department of Oncology and Pathology, Medical Radiobiology Section, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | |
Collapse
|
18
|
Zou L, Zhang PG, Zou S, Li Y, He Q. The Half-Life of Thymidine Kinase 1 in Serum Measured by ECL Dot Blot: A Potential Marker for Monitoring the Response to Surgery of Patients with Gastric Cancer. Int J Biol Markers 2018; 17:135-40. [PMID: 12113581 DOI: 10.1177/172460080201700210] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thymidine kinase 1 in serum (STK1) of patients with gastric cancer was determined by two methods: ECL dot blot and radioactivity assay. Both measurements showed significantly different values for preoperative STK1 and healthy STK1 (p=0.012 for ECL dot blot and p=0.003 for the radioactivity assay). The preliminary results of ECL dot blot STK1 measurement showed that in tumor-free subjects the level of the enzyme was significantly reduced to 52.7% 35 days after surgery (n=8, p=0.0106). The decrease in STK1 levels in the tumor-free subjects paralleled the decline of the half-life of the STK1 enzyme. In patients with distant metastases (n=6) the enzyme level had increased to 173% 35 days postoperatively. By contrast, with the radioactivity assay no significant differences in thymidine kinase activity for 0-day-postoperative patients and 35-day-postoperative tumor-free patients was found (p=0.329). The activity decreased to 80% in 35-day-postoperative patients with metastatic disease. We suggest that the value of the half-life of STK1 measured by ECL dot blot can be used as a potential marker for monitoring the response to surgery in patients with gastric or other cancers one month after surgery.
Collapse
Affiliation(s)
- L Zou
- Department of Surgery, Wuhan University Hospital, China
| | | | | | | | | |
Collapse
|
19
|
Irwin CR, Hitt MM, Evans DH. Targeting Nucleotide Biosynthesis: A Strategy for Improving the Oncolytic Potential of DNA Viruses. Front Oncol 2017; 7:229. [PMID: 29018771 PMCID: PMC5622948 DOI: 10.3389/fonc.2017.00229] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022] Open
Abstract
The rapid growth of tumors depends upon elevated levels of dNTPs, and while dNTP concentrations are tightly regulated in normal cells, this control is often lost in transformed cells. This feature of cancer cells has been used to advantage to develop oncolytic DNA viruses. DNA viruses employ many different mechanisms to increase dNTP levels in infected cells, because the low concentration of dNTPs found in non-cycling cells can inhibit virus replication. By disrupting the virus-encoded gene(s) that normally promote dNTP biosynthesis, one can assemble oncolytic versions of these agents that replicate selectively in cancer cells. This review covers the pathways involved in dNTP production, how they are dysregulated in cancer cells, and the various approaches that have been used to exploit this biology to improve the tumor specificity of oncolytic viruses. In particular, we compare and contrast the ways that the different types of oncolytic virus candidates can directly modulate these processes. We limit our review to the large DNA viruses that naturally encode homologs of the cellular enzymes that catalyze dNTP biogenesis. Lastly, we consider how this knowledge might guide future development of oncolytic viruses.
Collapse
Affiliation(s)
- Chad R Irwin
- Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Mary M Hitt
- Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.,Faculty of Medicine and Dentistry, Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - David H Evans
- Faculty of Medicine and Dentistry, Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.,Faculty of Medicine and Dentistry, Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Choi AH, O'Leary MP, Fong Y, Chen NG. From Benchtop to Bedside: A Review of Oncolytic Virotherapy. Biomedicines 2016; 4:biomedicines4030018. [PMID: 28536385 PMCID: PMC5344257 DOI: 10.3390/biomedicines4030018] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/14/2022] Open
Abstract
Oncolytic viruses (OVs) demonstrate the ability to replicate selectively in cancer cells, resulting in antitumor effects by a variety of mechanisms, including direct cell lysis and indirect cell death through immune-mediate host responses. Although the mechanisms of action of OVs are still not fully understood, major advances have been made in our understanding of how OVs function and interact with the host immune system, resulting in the recent FDA approval of the first OV for cancer therapy in the USA. This review provides an overview of the history of OVs, their selectivity for cancer cells, and their multifaceted mechanism of antitumor action, as well as strategies employed to augment selectivity and efficacy of OVs. OVs in combination with standard cancer therapies are also discussed, as well as a review of ongoing human clinical trials.
Collapse
Affiliation(s)
- Audrey H Choi
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Michael P O'Leary
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Nanhai G Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
- Center for Gene Therapy, Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
21
|
Abstract
Two different strategies have been developed for imaging the proliferative status of solid tumors with the functional imaging technique, Positron Emission Tomography (PET). The first strategy uses carbon-11 labeled thymidine and/or, more recently, fluorine-18 labeled thymidine analogs. These agents are a substrate for the enzyme thymidine kinase-1 (TK-1) and provide a pulse label of the number of cells in S phase. The second method for imaging the proliferative status of a tumor uses radiolabeled ligands that bind to the sigma-2 receptor which has a 10-fold higher density in proliferating (P) tumor cells versus quiescent (Q) tumor cells. This article compares and contrasts the two different strategies for imaging the proliferative status of solid tumors, and describes the strengths and weaknesses of each approach.
Collapse
|
22
|
Agarwal HK, Khalil A, Ishita K, Yang W, Nakkula RJ, Wu LC, Ali T, Tiwari R, Byun Y, Barth RF, Tjarks W. Synthesis and evaluation of thymidine kinase 1-targeting carboranyl pyrimidine nucleoside analogs for boron neutron capture therapy of cancer. Eur J Med Chem 2015; 100:197-209. [PMID: 26087030 DOI: 10.1016/j.ejmech.2015.05.042] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 05/24/2015] [Accepted: 05/26/2015] [Indexed: 10/23/2022]
Abstract
A library of sixteen 2nd generation amino- and amido-substituted carboranyl pyrimidine nucleoside analogs, designed as substrates and inhibitors of thymidine kinase 1 (TK1) for potential use in boron neutron capture therapy (BNCT) of cancer, was synthesized and evaluated in enzyme kinetic-, enzyme inhibition-, metabolomic-, and biodistribution studies. One of these 2nd generation carboranyl pyrimidine nucleoside analogs (YB18A [3]), having an amino group directly attached to a meta-carborane cage tethered via ethylene spacer to the 3-position of thymidine, was approximately 3-4 times superior as a substrate and inhibitor of hTK1 than N5-2OH (2), a 1st generation carboranyl pyrimidine nucleoside analog. Both 2 and 3 appeared to be 5'-monophosphorylated in TK1(+) RG2 cells, both in vitro and in vivo. Biodistribution studies in rats bearing intracerebral RG2 glioma resulted in selective tumor uptake of 3 with an intratumoral concentration that was approximately 4 times higher than that of 2. The obtained results significantly advance the understanding of the binding interactions between TK1 and carboranyl pyrimidine nucleoside analogs and will profoundly impact future design strategies for these agents.
Collapse
Affiliation(s)
- Hitesh K Agarwal
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Ahmed Khalil
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Keisuke Ishita
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Weilian Yang
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Robin J Nakkula
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Lai-Chu Wu
- Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Tehane Ali
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Rohit Tiwari
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA
| | - Youngjoo Byun
- College of Pharmacy, Korea University, Sejong, Republic of Korea
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| | - Werner Tjarks
- Division of Medicinal Chemistry & Pharmacognosy, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
23
|
Conrad SJ, El-Aswad M, Kurban E, Jeng D, Tripp BC, Nutting C, Eversole R, Mackenzie C, Essani K. Oncolytic tanapoxvirus expressing FliC causes regression of human colorectal cancer xenografts in nude mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:19. [PMID: 25887490 PMCID: PMC4337313 DOI: 10.1186/s13046-015-0131-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/29/2015] [Indexed: 12/12/2022]
Abstract
Colorectal cancers are significant causes of morbidity and mortality and existing therapies often perform poorly for individuals afflicted with advanced disease. Oncolytic virotherapy is an emerging therapeutic modality with great promise for addressing this medical need. Herein we describe the in vivo testing of recombinant variants of the tanapoxvirus (TPV). Recombinant viruses were made ablated for either the 66R gene (encoding a thymidine kinase), the 2L gene (encoding a TNF-binding protein), or both. Some of the recombinants were armed to express mouse chemotactic protein 1 (mCCL2/mMCP-1), mouse granulocyte-monocyte colony stimulating factor (mGM-CSF), or bacterial flagellin (FliC). Tumors were induced in athymic nude mice by implantation of HCT 116 cells and subsequently treated by a single intratumoral injection of one of the recombinant TPVs. Histological examination showed a common neoplastic cell type and a range of immune cell infiltration, necrosis, and tumor cell organization. Significant regression was seen in tumors treated with virus TPV/Δ2L/Δ66R/fliC, and to a lesser extent the recombinants TPV/Δ2L and TPV/Δ66R. Our results suggest that oncolytic recombinants of the TPV armed with activators of the innate immune response may be effective virotherapeutic agents for colorectal cancers in humans and should be explored further to fully realize their potential.
Collapse
Affiliation(s)
- Steven J Conrad
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Mohamed El-Aswad
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Esaw Kurban
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - David Jeng
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Brian C Tripp
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Charles Nutting
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Robert Eversole
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Charles Mackenzie
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA.
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| |
Collapse
|
24
|
Effects of CO2 pneumoperitoneum on the expression of thymidine kinase 1 and Ki67 in colorectal carcinoma cells. Surg Endosc 2014; 28:2863-70. [DOI: 10.1007/s00464-014-3539-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 03/10/2014] [Indexed: 12/13/2022]
|
25
|
Liu YP, Wang J, Avanzato VA, Bakkum-Gamez JN, Russell SJ, Bell JC, Peng KW. Oncolytic vaccinia virotherapy for endometrial cancer. Gynecol Oncol 2014; 132:722-9. [PMID: 24434058 PMCID: PMC3977925 DOI: 10.1016/j.ygyno.2014.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Oncolytic virotherapy is a promising modality in endometrial cancer (EC) therapy. In this study, we compared the efficacy of the Copenhagen and Wyeth strains of oncolytic vaccinia virus (VV) incorporating the human thyroidal sodium iodide symporter (hNIS) as a reporter gene (VVNIS-C and VVNIS-W) in EC. METHODS Infectivity of VVNIS-C and VVNIS-W in type I (HEC1A, Ishikawa, KLE, RL95-2, and AN3 CA) and type II (ARK-1, ARK-2, and SPEC-2) human EC cell lines was evaluated. Athymic mice with ARK-2 or AN3 CA xenografts were treated with one intravenous dose of VVNIS-C or VVNIS-W. Tumor regression and in vivo infectivity were monitored via NIS expression using SPECT-CT imaging. RESULTS All EC cell lines except KLE were susceptible to infection and killing by VVNIS-C and VVNIS-W in vitro. VVNIS-C had higher infectivity and oncolytic activity than VVNIS-W in all cell lines, most notably in AN3 CA. Intravenous VVNIS-C was more effective at controlling AN3 CA xenograft growth than VVNIS-W, while both VVNIS-C and VVNIS-W ceased tumor growth and induced tumor regression in 100% of mice bearing ARK-2 xenografts. CONCLUSION Overall, VVNIS-C has more potent oncolytic viral activity than VVSIN-W in EC. VV appears to be most active in type II EC. Novel therapies are needed for the highly lethal type II EC histologies and further development of a VV clinical trial in type II EC is warranted.
Collapse
Affiliation(s)
- Yu-Ping Liu
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON KlY 4E9, Canada
| | - Victoria A Avanzato
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Pennsylvania State University, State College, PA, USA
| | | | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON KlY 4E9, Canada
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
26
|
Dave RV, Jebar AHS, Jennings VA, Adair RA, West EJ, Errington-Mais F, Toogood GJ, Melcher AA. Viral warfare! Front-line defence and arming the immune system against cancer using oncolytic vaccinia and other viruses. Surgeon 2014; 12:210-20. [PMID: 24502935 DOI: 10.1016/j.surge.2014.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 12/26/2013] [Accepted: 01/03/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite mankind's many achievements, we are yet to find a cure for cancer. We are now approaching a new era which recognises the promise of harnessing the immune system for anti-cancer therapy. Pathogens have been implicated for decades as potential anti-cancer agents, but implementation into clinical therapy has been plagued with significant drawbacks. Newer 'designer' agents have addressed some of these concerns, in particular, a new breed of oncolytic virus: JX-594, a genetically engineered pox virus, is showing promise. OBJECTIVE To review the current literature on the use of oncolytic viruses in the treatment of cancer; both by direct oncolysis and stimulation of the immune system. The review will provide a background and historical progression for the surgeon on tumour immunology, and the interplay between oncolytic viruses, immune cells, inflammation on tumourigenesis. METHODS A literature review was performed using the Medline database. CONCLUSIONS Viral therapeutics hold promise as a novel treatment modality for the treatment of disseminated malignancy. It provides a multi-pronged attack against tumour burden; direct tumour cell lysis, exposure of tumour-associated antigens (TAA), induction of immune danger signals, and recognition by immune effector cells.
Collapse
Affiliation(s)
- R V Dave
- Department of Hepatobiliary Surgery, St James University Hospital, Leeds, UK; Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - A H S Jebar
- Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - V A Jennings
- Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - R A Adair
- Department of Hepatobiliary Surgery, St James University Hospital, Leeds, UK; Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - E J West
- Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - F Errington-Mais
- Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK
| | - G J Toogood
- Department of Hepatobiliary Surgery, St James University Hospital, Leeds, UK
| | - A A Melcher
- Targeted and Biological Therapies, Leeds Institute of Molecular Medicine, Leeds, UK.
| |
Collapse
|
27
|
de Almeida A, Oliveira BL, Correia JD, Soveral G, Casini A. Emerging protein targets for metal-based pharmaceutical agents: An update. Coord Chem Rev 2013. [DOI: 10.1016/j.ccr.2013.01.031] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
28
|
Abstract
Thymidine kinase 1 (TK 1-fetal) is a cell cycle-dependent marker that increases dramatically during the S-phase of the cell cycle. In this review, the authors discuss serum levels of thymidine kinase in a variety of neoplasias. Determination of thymidine kinase helps to monitor the follow-up of solid tumours and haematological malignancies as well as indicating the efficacy of adjuvant and palliative chemotherapy. Elevated levels of thymidine kinase must always be interpreted together with a detailed knowledge of the patient's condition because nonspecific elevations of serum levels (inflammatory and autoimmune diseases) must be excluded.
Collapse
Affiliation(s)
- Ondrej Topolcan
- Charles University Prague, Medical Faculty in Pilsen, Department of Nuclear Medicine, Faculty Hospital Pilsen, 13 Edwarda Benese, 305 99 Pilsen, Czech Republic +420 377402948 ; +420 377402454 ;
| | | |
Collapse
|
29
|
Design, synthesis, and in vitro and in vivo biological studies of a 3'-deoxythymidine conjugate that potentially kills cancer cells selectively. PLoS One 2012; 7:e52199. [PMID: 23300611 PMCID: PMC3530607 DOI: 10.1371/journal.pone.0052199] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 11/12/2012] [Indexed: 01/01/2023] Open
Abstract
Thymidine kinases (TKs) have been considered one of the potential targets for anticancer therapeutic because of their elevated expressions in cancer cells. However, nucleobase analogs targeting TKs have shown poor selective cytotoxicity in cancer cells despite effective antiviral activity. 3′-Deoxythymidine phenylquinoxaline conjugate (dT-QX) was designed as a novel nucleobase analog to target TKs in cancer cells and block cell replication via conjugated DNA intercalating quinoxaline moiety. In vitro cell screening showed that dT-QX selectively kills a variety of cancer cells including liver carcinoma, breast adenocarcinoma and brain glioma cells; whereas it had a low cytotoxicity in normal cells such as normal human liver cells. The anticancer activity of dT-QX was attributed to its selective inhibition of DNA synthesis resulting in extensive mitochondrial superoxide stress in cancer cells. We demonstrate that covalent linkage with 3′-deoxythymidine uniquely directed cytotoxic phenylquinoxaline moiety more toward cancer cells than normal cells. Preliminary mouse study with subcutaneous liver tumor model showed that dT-QX effectively inhibited the growth of tumors. dT-QX is the first molecule of its kind with highly amendable constituents that exhibits this selective cytotoxicity in cancer cells.
Collapse
|
30
|
Crosstalk between immune cell and oncolytic vaccinia therapy enhances tumor trafficking and antitumor effects. Mol Ther 2012; 21:620-8. [PMID: 23229093 DOI: 10.1038/mt.2012.257] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The combination of an oncolytic virus, that directly destroys tumor cells and mediates an acute immune response, with an immune cell therapy, capable of further enlisting and enhancing the host immune response, has the potential to create a potent therapeutic effect. We have previously developed several strategies for optimizing the delivery of oncolytic vaccinia virus vectors to their tumor targets, including the use of immune cell-based carrier vehicles and the incorporation of mutations that increase production of the enveloped form of vaccinia (extracellular enveloped viral (EEV)) that is better adapted to spread within a host. Here, we initially combine these approaches to create a novel therapeutic, consisting of an immune cell (cytokine-induced killer, CIK) preloaded with an oncolytic virus that is EEV enhanced. This resulted in direct interaction between the viral and immune cell components with each assisting the other in directing the therapy to the tumor and so enhancing the antitumor effects. This effect could be further improved through CCL5 expression from the virus. The resulting multicomponent therapy displays the ability for synergistic crosstalk between components, so significantly enhancing tumor trafficking and antitumor effects.
Collapse
|
31
|
Oncolytic viruses in the treatment of bladder cancer. Adv Urol 2012; 2012:404581. [PMID: 22899907 PMCID: PMC3414001 DOI: 10.1155/2012/404581] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/05/2012] [Indexed: 01/22/2023] Open
Abstract
Bladder carcinoma is the second most common malignancy of the urinary tract. Up to 85% of patients with bladder cancer are diagnosed with a tumor that is limited to the bladder mucosa (Ta, T1, and CIS). These stages are commonly termed as non-muscle-invasive bladder cancer (NMIBC). Although the treatment of NMIBC has greatly improved in recent years, there is a need for additional therapies when patients fail bacillus Calmette-Guérin (BCG) and chemotherapeutic agents. We propose that bladder cancer may be an ideal target for oncolytic viruses engineered to selectively replicate in and lyse tumor cells leaving normal cells unharmed. In support of this hypothesis, here we review current treatment strategies for bladder cancer and their shortcomings, as well as recent advancements in oncolytic viral therapy demonstrating encouraging safety profiles and antitumor activity.
Collapse
|
32
|
Thamm DH, Kamstock DA, Sharp CR, Johnson SI, Mazzaferro E, Herold LV, Barnes SM, Winkler K, Selting KA. Elevated serum thymidine kinase activity in canine splenic hemangiosarcoma*. Vet Comp Oncol 2011; 10:292-302. [PMID: 22236280 DOI: 10.1111/j.1476-5829.2011.00298.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thymidine kinase 1 (TK1) is a soluble biomarker associated with DNA synthesis. This prospective study evaluated serum TK1 activity in dogs presenting with hemoabdomen and a splenic mass. An ELISA using azidothymidine as a substrate was used to evaluate TK1 activity. Sixty-two dogs with hemoabdomen and 15 normal controls were studied. Serum TK1 activity was significantly higher in dogs with hemangiosarcoma (HSA) than in normal dogs (mean ± SEM = 17.0 ± 5.0 and 2.01 ± 0.6, respectively), but not dogs with benign disease (mean ± SEM = 10.0 ± 3.3). Using a cut-off of 6.55 U/L, TK activity demonstrated a sensitivity of 0.52, specificity of 0.93, positive predictive value of 0.94 and negative predictive value of 0.48 for distinguishing HSA versus normal. When interval thresholds of <1.55 and >7.95 U/L were used together, diagnostic utility was increased. Serum TK1 evaluation may help to discriminate between benign disease and HSA in dogs with hemoabdomen and a splenic mass.
Collapse
Affiliation(s)
- D H Thamm
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rueger MA, Ameli M, Li H, Winkeler A, Rueckriem B, Vollmar S, Galldiks N, Hesselmann V, Fraefel C, Wienhard K, Heiss WD, Jacobs AH. [18F]FLT PET for non-invasive monitoring of early response to gene therapy in experimental gliomas. Mol Imaging Biol 2011; 13:547-557. [PMID: 20563754 DOI: 10.1007/s11307-010-0361-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The purpose of this study was to investigate the potential of 3'-deoxy-3'-[¹⁸F]fluorothymidine ([¹⁸F]FLT) positron emission tomography (PET) to detect early treatment responses in gliomas. Human glioma cells were stably transduced with genes yielding therapeutic activity, sorted for different levels of exogenous gene expression, and implanted subcutaneously into nude mice. Multimodality imaging during prodrug therapy included (a) magnetic resonance imaging, (b) PET with 9-(4-[¹⁸F]fluoro-3-hydroxymethylbutyl)guanine assessing exogenous gene expression, and (c) repeat [¹⁸F]FLT PET assessing antiproliferative therapeutic response. All stably transduced gliomas responded to therapy with significant reduction in tumor volume and [¹⁸F]FLT accumulation within 3 days after initiation of therapy. The change in [¹⁸F]FLT uptake before and after treatment correlated to volumetrically calculated growth rates. Therapeutic efficacy as monitored by [¹⁸F]FLT PET correlated to levels of therapeutic gene expression measured in vivo. Thus, [¹⁸F]FLT PET assesses early antiproliferative effects, making it a promising radiotracer for the development of novel treatments for glioma.
Collapse
Affiliation(s)
- Maria A Rueger
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Center for Molecular Medicine (CMMC), Cologne, Germany.,Departments of Neurology, University Hospital Cologne, Cologne, Germany
| | - Mitra Ameli
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Departments of Neurology, University Hospital Cologne, Cologne, Germany
| | - Hongfeng Li
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Alexandra Winkeler
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany.,Center for Molecular Medicine (CMMC), Cologne, Germany
| | | | - Stefan Vollmar
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Norbert Galldiks
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Volker Hesselmann
- Department of Radiology, University Hospital Cologne, Cologne, Germany
| | - Cornel Fraefel
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Klaus Wienhard
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Wolf-Dieter Heiss
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany
| | - Andreas H Jacobs
- Laboratory for Gene Therapy and Molecular Imaging, Max Planck-Institute for Neurological Research, Gleuelerstr. 50, 50931, Cologne, Germany. .,Center for Molecular Medicine (CMMC), Cologne, Germany. .,European Institute for Molecular Imaging (EIMI), University of Münster, Münster, Germany.
| |
Collapse
|
34
|
Brockenbrough JS, Souquet T, Morihara JK, Stern JE, Hawes SE, Rasey JS, Leblond A, Wiens LW, Feng Q, Grierson J, Vesselle H. Tumor 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) uptake by PET correlates with thymidine kinase 1 expression: static and kinetic analysis of (18)F-FLT PET studies in lung tumors. J Nucl Med 2011; 52:1181-8. [PMID: 21764789 DOI: 10.2967/jnumed.111.089482] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED We report the first, to our knowledge, findings describing the relationships between both static and dynamic analysis parameters of 3'-deoxy-3'-(18)F-fluorothymidine ((18)F-FLT) PET and the expression of the proliferation marker Ki-67, and the protein expression and enzymatic activity of thymidine kinase-1 (TK1) in surgically resected lung lesions. METHODS Static and dynamic analyses (4 rate constants and 2 compartments) of (18)F-FLT PET images were performed in a cohort of 25 prospectively accrued, clinically suspected lung cancer patients before surgical resection (1 lesion was found to be benign after surgery). The maximal and overall averaged expression of Ki-67 and TK1 were determined by semiquantitative analysis of immunohistochemical staining. TK1 enzymatic activity was determined by in vitro assay of extracts prepared from flash-frozen samples of the same tumors. RESULTS Static (18)F-FLT uptake (partial-volume-corrected maximum-pixel standardized uptake value from 60- to 90-min summed dynamic data) was significantly correlated with the overall (ρ = 0.57, P = 0.006) and maximal (ρ = 0.69, P < 0.001) immunohistochemical expressions of Ki-67 and TK1 (overall expression: ρ = 0.65, P = 0.001; maximal expression: ρ = 0.68, P < 0.001) but not with TK1 enzymatic activity (ρ = 0.34, P = 0.146). TK1 activity was significantly correlated with TK1 protein expression only when immunohistochemistry was scored for maximal expression (ρ = 0.52, P = 0.029). Dynamic analysis of (18)F-FLT PET revealed correlations between the flux constant (K(FLT)) and both overall (ρ = 0.53, P = 0.014) and maximal (ρ = 0.50, P = 0.020) TK1 protein expression. K(FLT) was also associated with both overall (ρ = 0.59, P = 0.005) and maximal (ρ = 0.63, P = 0.002) Ki-67 expression. We observed no significant correlations between TK1 enzyme activity and K(FLT). In addition, no significant relationships were found between TK1 expression, TK1 activity, or Ki-67 expression and any of the compartmental rate constants. CONCLUSION The absence of observable correlations of the imaging parameters with TK1 activity suggests that (18)F-FLT uptake and retention within cells may be complicated by a variety of still undetermined factors in addition to TK1 enzymatic activity.
Collapse
Affiliation(s)
- J Scott Brockenbrough
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA 98195-7115, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Mazouni C, Fina F, Romain S, Ouafik L, Bonnier P, Brandone JM, Martin PM. Epstein-Barr virus as a marker of biological aggressiveness in breast cancer. Br J Cancer 2010; 104:332-7. [PMID: 21179039 PMCID: PMC3031896 DOI: 10.1038/sj.bjc.6606048] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose: Although a potential role of the Epstein-Barr virus (EBV) in the pathogenesis of breast cancer (BC) has been underlined, results remain conflicting. Particularly, the impact of EBV infection on biological markers of BC has received little investigation. Methods: In this study, we established the frequency of EBV-infected BC using real-time quantitative PCR (RT–PCR) in 196 BC specimens. Biological and pathological characteristics according to EBV status were evaluated. Results: EBV DNA was present in 65 of the 196 (33.2%) cases studied. EBV-positive BCs tended to be tumours with a more aggressive phenotype, more frequently oestrogen receptor negative (P=0.05) and with high histological grade (P=0.01). Overexpression of thymidine kinase activity was higher in EBV-infected BC (P=0.007). The presence of EBV was weakly associated with HER2 gene amplification (P=0.08). Conclusion: Our study provides evidence for EBV-associated BC undergoing distinct carcinogenic processes, with more aggressive features.
Collapse
Affiliation(s)
- C Mazouni
- Laboratoire de transfert d'oncologie biologique, Assistance Publique - Hôpitaux de Marseille, Faculté de Médecine Nord, Marseille, France.
| | | | | | | | | | | | | |
Collapse
|
36
|
von Euler H, Eriksson S. Comparative aspects of the proliferation marker thymidine kinase 1 in human and canine tumour diseases. Vet Comp Oncol 2010; 9:1-15. [PMID: 21303450 DOI: 10.1111/j.1476-5829.2010.00238.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
As cell proliferation is one of the hallmarks of cancer, various types of proliferation markers are used as important tools in diagnosis, prognosis, treatment decision-making and follow-up in clinical oncology. The S phase-specific protein thymidine kinase 1 (TK1) can be used in immunohistochemistry for RNA/protein expression in tissue specimens and for activity or protein/peptide levels in serum from patients. TK1 has been used mainly in haematologic malignancies in humans, but also found beneficial in canine malignancies. As the protein sequence homology is high between humans and dogs, findings in canine models will have a high comparative value in further human research as well. In the present review, we will focus on the recent results concerning TK1's S phase-correlated expression, increased serum levels of TK1 in patients with malignancies and the relevance for veterinary and comparative oncology. Finally, the benefit of recently developed specific anti-TK1 antibodies suitable for immunologic assay is discussed.
Collapse
Affiliation(s)
- H von Euler
- Center of Clinical Comparative Oncology (C3O), Department of Clinical Sciences, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden.
| | | |
Collapse
|
37
|
Bartholomä M, Vortherms A, Hillier S, Ploier B, Joyal J, Babich J, Doyle R, Zubieta J. Synthesis, Cytotoxicity, and Insight into the Mode of Action of Re(CO)3 Thymidine Complexes. ChemMedChem 2010; 5:1513-29. [DOI: 10.1002/cmdc.201000196] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
38
|
Li Z, Wang Y, He J, Ma J, Zhao L, Chen H, Li N, Zhou J, He E, Skog S. Serological thymidine kinase 1 is a prognostic factor in oesophageal, cardial and lung carcinomas. Eur J Cancer Prev 2010; 19:313-318. [PMID: 20479645 DOI: 10.1097/cej.0b013e32833ad320] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this study we examine the use of the concentration of thymidine kinase 1 in serum (STK1) as a prognostic factor in routine clinical settings. For this purpose we used sera from patients with oesophageal (n=101) and cardial (n=39) carcinomas and nonsmall-cell lung carcinoma (NSCLC) (n=157). Sera from healthy individuals (n=95) were used as controls. STK1 was analysed by a chemiluminiscence dot blot assay. The mean STK1 concentrations and the STK1 positive rates of the patients with oesophageal and cardial carcinomas and with NSCLC were significantly higher as compared with healthy controls (P=0.01). The mean STK1 value of oesophageal carcinoma patients correlated with T-values (P=0.021) and with stage (P<0.005), but not with grade. The mean STK1 value of cardial carcinoma patients did not correlate with grade. No data on stage and T-values were available for these patients, due to advanced disease. The mean STK1 value of NSCLC patients with squamous cell carcinoma was significantly higher as compared with adenocarcinoma type (P=0.024). The mean STK1 value of the NSCLC patients correlated with clinical grade (P=0.006), T-values (P=0.001), stage (P=0.035) and to size of the tumour (P=0.030). The mean STK1 value and the number of STK1 positive patients were also higher in recurrent NSCLC patients. There was a tendency that stage I-II NSCLC patients with an STK1 level above 2 pmol/l showed a higher frequency of recurrence/death than patients below 1 pmol/l. Our results show that STK1 is a useful marker for prognosis in patients with oesophageal, cardial and lung carcinomas.
Collapse
Affiliation(s)
- Zhishan Li
- Department of Thoracic Surgery Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Science, Beijing, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Despite the recognized limitations of (18)Fluorodeoxyglucose positron emission tomography (FDG-PET) in brain tumor imaging due to the high background of normal gray matter, this imaging modality provides critical information for the management of patients with cerebral neoplasms with regard to the following aspects: (1) providing a global picture of the tumor and thus guiding the appropriate site for stereotactic biopsy, and thereby enhancing its accuracy and reducing the number of biopsy samples; and (2) prediction of biologic behavior and aggressiveness of the tumor, thereby aiding in prognosis. Another area, which has been investigated extensively, includes differentiating recurrent tumor from treatment-related changes (eg, radiation necrosis and postsurgical changes). Furthermore, FDG-PET has demonstrated its usefulness in differentiating lymphoma from toxoplasmosis in patients with acquired immune deficiency syndrome with great accuracy, and is used as the investigation of choice in this setting. Image coregistration with magnetic resonance imaging and delayed FDG-PET imaging are 2 maneuvers that substantially improve the accuracy of interpretation, and hence should be routinely employed in clinical settings. In recent years an increasing number of brain tumor PET studies has used other tracers (like labeled methionine, tyrosine, thymidine, choline, fluoromisonidazole, EF5, and so forth), of which positron-labeled amino acid analogues, nucleotide analogues, and the hypoxia imaging tracers are of special interest. The major advantage of these radiotracers over FDG is the markedly lower background activity in normal brain tissue, which allows detection of small lesions and low-grade tumors. The promise of the amino acid PET tracers has been emphasized due to their higher sensitivity in imaging recurrent tumors (particularly the low-grade ones) and better accuracy for differentiating between recurrent tumors and treatment-related changes compared with FDG. The newer PET tracers have also shown great potential to image important aspects of tumor biology and thereby demonstrate ability to forecast prognosis. The value of hypoxia imaging tracers (such as fluoromisonidazole or more recently EF5) is substantial in radiotherapy planning and predicting treatment response. In addition, they may play an important role in the future in directing and monitoring targeted hypoxic therapy for tumors with hypoxia. Development of optimal image segmentation strategy with novel PET tracers and multimodality imaging is an approach that deserves mention in the era of intensity modulated radiotherapy, and which is likely to have important clinical and research applications in radiotherapy planning in patients with brain tumor.
Collapse
Affiliation(s)
- Sandip Basu
- Radiation Medicine Centre (BARC), Tata Memorial Hospital Annexe, Parel, Bombay 400012, India
| | | |
Collapse
|
40
|
Alberto R, N’Dongo HP, Clericuzio M, Bonetti S, Gabano E, Cassino C, Ravera M, Osella D. Functionalized thymidine derivatives as carriers for the γ-emitter technetium tricarbonyl moiety. Inorganica Chim Acta 2009. [DOI: 10.1016/j.ica.2009.06.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
41
|
Thymidine kinase 1 expression defines an activated G1 state of the cell cycle as revealed with site-specific antibodies and ArrayScan assays. Eur J Cell Biol 2009; 88:779-85. [PMID: 19726104 DOI: 10.1016/j.ejcb.2009.06.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Revised: 06/18/2009] [Accepted: 06/29/2009] [Indexed: 11/21/2022] Open
Abstract
Thymidine kinase 1 (TK1) is a DNA salvage enzyme involved in the synthesis of thymidine triphosphate needed during S phase. Although TK1 has been utilized as a cell proliferation marker for many years no well-characterized antibodies are available. The preparation and properties of two types of poly- and monoclonal anti-TK1 peptide antibodies are described and they are used to determine the levels of TK1 in intact cells. Expression of TK1, c-fos, cyclin B1, Ki67, phosphorylated histone H3, phosphorylated ribosomal protein S6, as well as bromodeoxyuridine (BrdU) incorporation in human normal dermal fibroblast cultures were studied with high-content ArrayScan fluorescence microscopy. The levels of TK1 increased 6-7h after serum re-addition to starved cells as they passed through G1, S and G2/M phases, which was earlier than the increase in Ki67 protein levels and before BrdU incorporation was detected. Thus, a population of activated G1 cells with high TK1 and low Ki67 expression could be identified and their role in cell proliferation can now be clarified.
Collapse
|
42
|
Brockenbrough JS, Morihara JK, Hawes SE, Stern JE, Rasey JS, Wiens LW, Feng Q, Vesselle H. Thymidine kinase 1 and thymidine phosphorylase expression in non-small-cell lung carcinoma in relation to angiogenesis and proliferation. J Histochem Cytochem 2009; 57:1087-97. [PMID: 19654105 DOI: 10.1369/jhc.2009.952804] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The thymidine salvage pathway enzymes thymidine kinase 1 (TK1) and thymidine phosphorylase (TP) compete for thymidine as a substrate and catalyze opposing synthetic and catabolic reactions that have been implicated in the control of proliferation and angiogenesis, respectively. We investigated the relationship between the expression of TK1 and TP as they relate to proliferation (Ki-67 labeling index) and angiogenesis (Chalkley count of CD31-stained blood vessels) in a series of 110 non-small-cell lung cancer (NSCLC) tumors from patients prospectively enrolled in an imaging trial. TK1 and TP exhibited similar patterns of immunohistochemical distribution, in that each was found in both the nucleus and the cytoplasm of tumor cells. Each enzyme exhibited a significant positive correlation between its levels of nuclear and cytoplasmic expression. A significant positive correlation between TK1 expression and the Ki-67 labeling index (r = 0.53, p<0.001) was observed. TP was significantly positively correlated with Chalkley scoring of CD31 staining in high vs low Chalkley scoring samples (mean TP staining of 115.8 vs 79.9 scoring units, p<0.001), respectively. We did not observe a substantial inverse correlation between the TP and TK1 expression levels in the nuclear compartment (r = -0.17, p=0.08). Tumor size was not found to be associated with TK1, TP, Ki-67, or Chalkley score. These findings provide additional evidence for the role of thymidine metabolism in the complex interaction of proliferation and angiogenesis in NSCLC.
Collapse
Affiliation(s)
- J Scott Brockenbrough
- , Division of Nuclear Medicine, Department of Radiology, University of Washington Medical Center, Seattle, WA 98195-7115, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Kirn DH, Thorne SH. Targeted and armed oncolytic poxviruses: a novel multi-mechanistic therapeutic class for cancer. Nat Rev Cancer 2009; 9:64-71. [PMID: 19104515 DOI: 10.1038/nrc2545] [Citation(s) in RCA: 302] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Viruses have been engineered for cancer therapy in a variety of ways. Approaches include non-replicating gene therapy vectors, cancer vaccines and oncolytic viruses, but the clinical efficacy of these approaches has been limited by multiple factors. However, a new therapeutic class of oncolytic poxviruses has recently been developed that combines targeted and armed approaches for treating cancer. Initial preclinical and clinical results show that products from this therapeutic class can systemically target cancers in a highly selective and potent fashion using a multi-pronged mechanism of action.
Collapse
Affiliation(s)
- David H Kirn
- Jennerex Biotherapeutics Inc., San Francisco, California 94105, USA .
| | | |
Collapse
|
44
|
Desbouis D, Struthers H, Spiwok V, Küster T, Schibli R. Synthesis, In Vitro, and In Silico Evaluation of Organometallic Technetium and Rhenium Thymidine Complexes with Retained Substrate Activity toward Human Thymidine Kinase Type 1. J Med Chem 2008; 51:6689-98. [PMID: 18837546 DOI: 10.1021/jm800530p] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dominique Desbouis
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland, Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland, Department of Structure and Function of Saccharides, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 84538 Bratislava, Slovak Republic
| | - Harriet Struthers
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland, Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland, Department of Structure and Function of Saccharides, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 84538 Bratislava, Slovak Republic
| | - Vojtech Spiwok
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland, Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland, Department of Structure and Function of Saccharides, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 84538 Bratislava, Slovak Republic
| | - Tatiana Küster
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland, Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland, Department of Structure and Function of Saccharides, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 84538 Bratislava, Slovak Republic
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland, Center for Radiopharmaceutical Science, ETH-PSI-USZ, Paul Scherrer Institute, 5232 Villigen PSI, Switzerland, Department of Structure and Function of Saccharides, Institute of Chemistry, Slovak Academy of Sciences, Dubravska Cesta 9, 84538 Bratislava, Slovak Republic
| |
Collapse
|
45
|
The Targeted Oncolytic Poxvirus JX-594 Demonstrates Antitumoral, Antivascular, and Anti-HBV Activities in Patients With Hepatocellular Carcinoma. Mol Ther 2008; 16:1637-42. [DOI: 10.1038/mt.2008.143] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
46
|
Goldberg MF, Chawla S, Alavi A, Torigian DA, Melhem ER. PET and MR Imaging of Brain Tumors. PET Clin 2008; 3:293-315. [DOI: 10.1016/j.cpet.2009.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
47
|
Park BH, Hwang T, Liu TC, Sze DY, Kim JS, Kwon HC, Oh SY, Han SY, Yoon JH, Hong SH, Moon A, Speth K, Park C, Ahn YJ, Daneshmand M, Rhee BG, Pinedo HM, Bell JC, Kirn DH. Use of a targeted oncolytic poxvirus, JX-594, in patients with refractory primary or metastatic liver cancer: a phase I trial. Lancet Oncol 2008; 9:533-42. [DOI: 10.1016/s1470-2045(08)70107-4] [Citation(s) in RCA: 395] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
48
|
Chandra S, Tjarks W, Lorey DR, Barth RF. Quantitative subcellular imaging of boron compounds in individual mitotic and interphase human glioblastoma cells with imaging secondary ion mass spectrometry (SIMS). J Microsc 2008; 229:92-103. [PMID: 18173648 DOI: 10.1111/j.1365-2818.2007.01869.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Boron measurements at subcellular scale are essential in boron neutron capture therapy (BNCT) of cancer as the nuclear localization of boron-10 atoms can enhance the effectiveness of killing individual tumour cells. Since tumours contain a heterogeneous population of cells in interphase as well as in the M phase (mitotic division) of the cell cycle, it is important to evaluate the subcellular distribution of boron in both phases. In this work, the secondary ion mass spectrometry (SIMS) based imaging technique of ion microscopy was used to quantitatively image boron from two BNCT agents, clinically used p-boronophenylalanine (BPA) and 3-[4-(o-carboran-1-yl)butyl]thymidine (N4), in mitotic metaphase and interphase human glioblastoma T98G cells. N4 belongs to a class of experimental BNCT agents, designated 3-carboranyl thymidine analogues (3CTAs), which presumably accumulate selectively in cancer cells due to a process referred to as kinase-mediated trapping (KMT). The cells were exposed to BPA for 1 h and N4 for 2 h. A CAMECA IMS-3f SIMS ion microscope instrument capable of producing isotopic images with 500 nm spatial resolution was used in the study. Observations were made in cryogenically prepared fast frozen, and freeze-fractured, freeze-dried cells. Three discernible subcellular regions were studied: the nucleus, a characteristic mitochondria-rich perinuclear cytoplasmic region, and the remaining cytoplasm in interphase T98G cells. In metaphase cells, the chromosomes and the cytoplasm were studied for boron localization. Intracellular concentrations of potassium and sodium also were measured in each cell in which the subcellular boron concentrations were imaged. Since the healthy cells maintain a K/Na ratio of approximately 10 due to the presence of Na-K-ATPase in the plasma membrane of mammalian cells, these measurements provided validation for cryogenic sample preparation and indicated the analysis healthy, well preserved cells. The BPA-treated interphase cells revealed significantly lower concentrations of boron in the perinuclear mitochondria-rich cytoplasmic region as compared to the remaining cytoplasm and the nucleus, which were not significantly different from each other. In contrast, the BPA-treated metaphase cells revealed significantly lower concentration of boron in their chromosomes than cytoplasm. In addition, the cytoplasm of metaphase cells contained significantly less boron than the cytoplasm of interphase cells. These observations provide valuable information on the reduced uptake of boron from BPA in mitotic cells for BPA-mediated BNCT. SIMS observations on N4 revealed that boron was distributed throughout the interphase and mitotic cells, including the chromosomes. The presence of boron in chromosomes of metaphase cells treated with N4 is indicative of a possible incorporation of this thymidine analogue into DNA. The 3-D SIMS imaging approach for the analysis of mitotic cells shown in this work should be equally feasible to the evaluation of other BNCT agents.
Collapse
Affiliation(s)
- S Chandra
- Cornell SIMS Laboratory, Department of Earth and Atmospheric Sciences, Cornell University, Ithaca, NY, USA.
| | | | | | | |
Collapse
|
49
|
Thorne SH, Hwang THH, O’Gorman WE, Bartlett DL, Sei S, Kanji F, Brown C, Werier J, Cho JH, Lee DE, Wang Y, Bell J, Kirn DH. Rational strain selection and engineering creates a broad-spectrum, systemically effective oncolytic poxvirus, JX-963. J Clin Invest 2007; 117:3350-8. [PMID: 17965776 PMCID: PMC2040321 DOI: 10.1172/jci32727] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Accepted: 08/15/2007] [Indexed: 12/11/2022] Open
Abstract
Replication-selective oncolytic viruses (virotherapeutics) are being developed as novel cancer therapies with unique mechanisms of action, but limitations in i.v. delivery to tumors and systemic efficacy have highlighted the need for improved agents for this therapeutic class to realize its potential. Here we describe the rational, stepwise design and evaluation of a systemically effective virotherapeutic (JX-963). We first identified a highly potent poxvirus strain that also trafficked efficiently to human tumors after i.v. administration. This strain was then engineered to target cancer cells with activation of the transcription factor E2F and the EGFR pathway by deletion of the thymidine kinase and vaccinia growth factor genes. For induction of tumor-specific cytotoxic T lymphocytes, we further engineered the virus to express human GM-CSF. JX-963 was more potent than the previously used virotherapeutic Onyx-015 adenovirus and as potent as wild-type vaccinia in all cancer cell lines tested. Significant cancer selectivity of JX-963 was demonstrated in vitro in human tumor cell lines, in vivo in tumor-bearing rabbits, and in primary human surgical samples ex vivo. Intravenous administration led to systemic efficacy against both primary carcinomas and widespread organ-based metastases in immunocompetent mice and rabbits. JX-963 therefore holds promise as a rationally designed, targeted virotherapeutic for the systemic treatment of cancer in humans and warrants clinical testing.
Collapse
Affiliation(s)
- Steve H. Thorne
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Tae-Ho H. Hwang
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - William E. O’Gorman
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - David L. Bartlett
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Shizuko Sei
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Femina Kanji
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Christopher Brown
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Joel Werier
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Jin-Han Cho
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Dong-Ewon Lee
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - Yaohe Wang
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - John Bell
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| | - David H. Kirn
- Department of Pediatrics and Bio-X Program, James H. Clark Center, Stanford University School of Medicine, Stanford, California, USA.
Jennerex Biotherapeutics, San Francisco, California, USA.
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA.
Division of Surgical Oncology, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
Viral Vector Toxicology Section, LHTP, SAIC-Frederick Inc., National Cancer Institute–Frederick, Frederick, Maryland, USA.
Ottawa Health Research Institute, Ottawa, Ontario, Canada.
Biochemistry, Microbiology and Immunology Department, University of Ottawa, Ottawa, Ontario, Canada.
Ottawa General Hospital, Ottawa, Ontario, Canada.
Department of Radiology and
Department of Pharmacology, Medical College of Dong-A University, Busan, Republic of Korea.
Barts and the London School of Medicine and Dentistry, Queen Mary’s University of London, London, United Kingdom
| |
Collapse
|
50
|
Salskov A, Tammisetti VS, Grierson J, Vesselle H. FLT: Measuring Tumor Cell Proliferation In Vivo With Positron Emission Tomography and 3′-Deoxy-3′-[18F]Fluorothymidine. Semin Nucl Med 2007; 37:429-39. [PMID: 17920350 DOI: 10.1053/j.semnuclmed.2007.08.001] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|