1
|
Oncolytic Viruses and Hematological Malignancies: A New Class of Immunotherapy Drugs. ACTA ACUST UNITED AC 2020; 28:159-183. [PMID: 33704184 PMCID: PMC7816176 DOI: 10.3390/curroncol28010019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023]
Abstract
The use of viruses for tumour treatment has been imagined more than one hundred years ago, when it was reported that viral diseases were occasionally leading to a decrease in neoplastic lesions. Oncolytic viruses (OVs) seem to have a specific tropism for tumour cells. Previously, it was hypothesised that OVs’ antineoplastic actions were mainly due to their ability to contaminate, proliferate and destroy tumour cells and the immediate destructive effect on cells was believed to be the single mechanism of action of OVs’ action. Instead, it has been established that oncolytic viruses operate via a multiplicity of systems, including mutation of tumour milieu and a composite change of the activity of immune effectors. Oncolytic viruses redesign the tumour environment towards an antitumour milieu. The aim of our work is to evaluate the findings present in the literature about the use of OVs in the cure of haematological neoplastic pathologies such as multiple myeloma, acute and chronic myeloid leukaemia, and lymphoproliferative diseases. Further experimentations are essential to recognize the most efficient virus or treatment combinations for specific haematological diseases, and the combinations able to induce the strongest immune response.
Collapse
|
2
|
Jiang J, Cole D, Westwood N, Macpherson L, Farzaneh F, Mufti G, Tavassoli M, Gäken J. Crucial roles for protein kinase C isoforms in tumor-specific killing by apoptin. Cancer Res 2010; 70:7242-52. [PMID: 20719884 DOI: 10.1158/0008-5472.can-10-1204] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The chicken anemia virus-derived protein apoptin induces apoptosis in a variety of human malignant and transformed cells but not in normal cells. However, the mechanisms through which apoptin achieves its selective killing effects are not well understood. We developed a lentiviral vector encoding a green fluorescent protein-apoptin fusion gene (LV-GFP-AP) that can efficiently deliver apoptin into hematopoietic cells. Apoptin selectively killed the human multiple myeloma cell lines MM1.R and MM1.S, and the leukemia cell lines K562, HL60, U937, KG1, and NB4. In contrast, normal CD34(+) cells were not killed and maintained their differentiation potential in multilineage colony formation assays. In addition, dexamethasone-resistant MM1.R cells were found to be more susceptible to apoptin-induced cell death than the parental matched MM1.S cells. Death susceptibility correlated with increased phosphorylation and activation of the apoptin protein in MM1.R cells. Expression array profiling identified differential kinase profiles between MM1.R and MM1.S cells. Among these kinases, protein kinase Cβ (PKCβ) was found by immunoprecipitation and in vitro kinase studies to be a candidate kinase responsible for apoptin phosphorylation. Indeed, shRNA knockdown or drug-mediated inhibition of PKCβ significantly reduced apoptin phosphorylation. Furthermore, apoptin-mediated cell death proceeded through the upregulation of PKCβ, activation of caspase-9/3, cleavage of the PKCδ catalytic domain, and downregulation of the MERTK and AKT kinases. Collectively, these results elucidate a novel pathway for apoptin activation involving PKCβ and PKCδ. Further, they highlight the potential of apoptin and its cellular regulators to purge bone marrow used in autologous transplantation for multiple myeloma.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Haematological and Molecular Medicine, The Rayne Institute, King's College London, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Therapeutic Nucleic Acids. Gene Ther 2010. [DOI: 10.1007/978-88-470-1643-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
4
|
Jiang S, Xie Q, Zhang W, Zhou XQ, Jin YX. Preparation of anti-mouse caspase-12 mRNA hammerhead ribozyme and identification of its activity in vitro. World J Gastroenterol 2005; 11:4094-7. [PMID: 15996037 PMCID: PMC4502108 DOI: 10.3748/wjg.v11.i26.4094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To prepare and identify specific anti-mouse caspase-12 hammerhead ribozymes in vitro, in order to select a more effective ribozyme against mouse caspase-12 as a potential tool to rescue cells from endoplasmic reticulum stress induced apoptosis.
METHODS: Two hammerhead ribozymes directed separately against 138 and 218 site of nucleotide of mouse caspase-12 mRNA were designed by computer software, and their DNA sequences were synthesized. The synthesized ribozymes were cloned into an eukaryotic expression vector-neorpBSKU6 and embedded in U6 SnRNA context for further study. Mouse caspase-12 gene segment was cloned into PGEM-T vector under the control of T7 RNA polymerase promoter (containing gene sequence from positions nt 41 to nt 894) as target. In vitro transcription both the ribozymes and target utilize T7 promoter. The target was labeled with [α-32P]UTP, while ribozymes were not labeled. After gel purification the RNAs were dissolved in RNase free water. Ribozyme and target were incubated for 90 min at 37°C in reaction buffer (40 mmol/L Tris-HCL, pH 7.5, 10 mmol/L Mg2+). Molar ratio of ribozyme vs target was 30:1. Samples were analyzed on 6% PAGE (containing 8 mol/L urea).
RESULTS: Both caspase-12 and ribozyme gene sequences were successfully cloned into expression vector confirmed by sequencing. Ribozymes and caspase-12 mRNA were obtained by in vitro transcription. Cleavage experiment showed that in a physiological similar condition (37°C, pH 7.5), Rz138 and Rz218 both cleaved targets at predicted sites, for Rz138 the cleavage efficiency was about 100%, for Rz218 the value was 36.66%.
CONCLUSION: Rz138 prepared in vitro can site specific cleave mouse caspase-12 mRNA with an excellent efficiency. It shows a potential to suppress the expression of caspase-12 in vivo, thus provided a new way to protect cells from ER stress induced apoptosis.
Collapse
Affiliation(s)
- Shan Jiang
- Department of Infectious Disease, Ruijin Hospital, Shanghai Second Medical University, Shanghai 200025, China
| | | | | | | | | |
Collapse
|
5
|
Wiernik PH. Plasma cell myeloma and leukemia. ACTA ACUST UNITED AC 2004; 21:365-97. [PMID: 15338756 DOI: 10.1016/s0921-4410(03)21019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Affiliation(s)
- Peter H Wiernik
- Our Lady of Mercy Cancer Center, New York Medical College, Bronx 10466, USA.
| |
Collapse
|
6
|
Liu JH, Wu SC, Chen WS, Yen CC, Yang MH, Tsai YC, Chen PM. An intron promotes the anti-bcr-abl activities of a retrovirally expressed ribozyme in chronic myeloid leukemia cells. Biochem Biophys Res Commun 2004; 318:764-72. [PMID: 15144904 DOI: 10.1016/j.bbrc.2004.04.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Indexed: 11/29/2022]
Abstract
Ribozymes have been developed to cleave the bcr-abl transcripts and thereby suppress transforming activities of chronic myelogenous leukemia (CML) cells. However, the intracellular efficacy of vector-dependent ribozymes usually depends in part on their expression cassettes, which may affect their intracellular trafficking and distribution. In order to test effects of an intron in pre-fusion ribozyme on the anti-bcr-abl activities in CML cells, retroviral vectors harboring ribozyme expression cassettes with (RzI) or without (Rz) an intron-encoding sequence were used to transduce K562 cells. In terms of both reduction of the target bcr-abl mRNA and suppression of colony formation in soft agar and xenograft growth on SCID mice, the anti-bcr-abl efficacy of the RzI fusion ribozyme was significantly superior to that of Rz. These results also correlate with more cytoplasmic accumulation of the RzI fusion ribozymes than that of the Rz. This study suggests activities of a RNA polymerase II-driven fusion ribozyme against its targeted spliced mRNA are improved by incorporating an intron in its pre-splicing transcript. Noticeably, the improvement is contributed in part by subcellular co-localization.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Count
- Cytoplasm/metabolism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/biosynthesis
- Fusion Proteins, bcr-abl/genetics
- Gene Dosage
- Genetic Vectors/genetics
- Genetic Vectors/pharmacology
- Humans
- Introns/genetics
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Mice
- Mice, SCID
- Molecular Sequence Data
- RNA, Catalytic/biosynthesis
- RNA, Catalytic/genetics
- RNA, Catalytic/metabolism
- Retroviridae/genetics
- Time Factors
- Transcription, Genetic
- Transduction, Genetic
Collapse
Affiliation(s)
- Jin-Hwang Liu
- Division of Medical Oncology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei, Taiwan, ROC.
| | | | | | | | | | | | | |
Collapse
|
7
|
Li MJ, McMahon R, Snyder DS, Yee JK, Rossi JJ. Specific killing of Ph+ chronic myeloid leukemia cells by a lentiviral vector-delivered anti-bcr/abl small hairpin RNA. Oligonucleotides 2004; 13:401-9. [PMID: 15000831 DOI: 10.1089/154545703322617087] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Chronic myeloid leukemia (CML) is characterized by a reciprocal chromosomal translocation between chromosomes 9 and 22 t(9;22)(q34;q11) that causes fusion of the bcr and abl genes. Transcription and splicing of the fusion gene generate two major splice variants of the bcr/abl transcript that encode an oncoprotein with tyrosine kinase activity. We have taken advantage of lentiviral vectormediated delivery of anti-bcr/abl short hairpin RNAs (shRNA) to downregulate the bcr/abl transcript in Philadelphia chromosome-positive (Ph+) K562 leukemia cells. This downregulation caused complete inhibition of proliferation of these cells and was accompanied by >90% inhibition of the bcr/abl transcript and p210 protein. These results demonstrate the feasibility of using a lentiviral vector to stably transduce therapeutic shRNAs into leukemia cells for the potential ex vivo purging of Ph+ cells in an autologous hematopoietic cell transplant setting. Furthermore, the robust expression of the shRNAs from our lentiviral vector suggests that this system could be generally useful for the expression of other shRNAs.
Collapse
MESH Headings
- Base Sequence
- Cell Division
- Fusion Proteins, bcr-abl/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Gene Silencing
- Genetic Vectors
- Humans
- K562 Cells
- Lentivirus/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Philadelphia Chromosome
- Plasmids/genetics
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Ming-Jie Li
- Division of Molecular Biology, City of Hope Medical Center and Beckman Research Institute, Duarte, CA 91010, USA
| | | | | | | | | |
Collapse
|
8
|
Ciafrè SA, Niola F, Wannenes F, Farace MG. An anti-VEGF ribozyme embedded within the adenoviral VAI sequence inhibits glioblastoma cell angiogenic potential in vitro. J Vasc Res 2004; 41:220-8. [PMID: 15084786 DOI: 10.1159/000077777] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Accepted: 03/01/2004] [Indexed: 12/20/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) plays an important role in tumor angiogenesis, where it functions as one of the major angiogenic factors sustaining growth and draining catabolites. In this study, we developed an anti-VEGF ribozyme targeted to the 5' part of human VEGF mRNA. We endowed this ribozyme with an additional feature expected to improve its activity in vivo, by cloning it into a VAI transcriptional cassette. VAI is originally part of the adenovirus genome, and is characterized by high transcription rates, good stability due to its strong secondary structure and cytoplasmic localization. Transfection of U87 human glioblastoma cells with plasmid vectors encoding for this ribozyme resulted in a strong (-56%) reduction of VEGF secreted in the extracellular medium, indicating a good biological activity of the ribozyme. Moreover, this reduction in VEGF secretion had the important functional consequence of drastically diminishing the formation of tube-like structures of human umbilical vascular endothelial cells in a Matrigel in vitro angiogenesis assay. In conclusion, our VAI-embedded anti-VEGF ribozyme is a good inhibitor of angiogenesis in vitro, in a glioblastoma cell context. Thus, it may represent a useful tool for future applications in vivo, for antiangiogenic gene therapy of glioblastoma and of highly vascularized tumors.
Collapse
Affiliation(s)
- Silvia Anna Ciafrè
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Rome, Italy.
| | | | | | | |
Collapse
|
9
|
Pennati M, Binda M, Colella G, Zoppe' M, Folini M, Vignati S, Valentini A, Citti L, De Cesare M, Pratesi G, Giacca M, Daidone MG, Zaffaroni N. Ribozyme-mediated inhibition of survivin expression increases spontaneous and drug-induced apoptosis and decreases the tumorigenic potential of human prostate cancer cells. Oncogene 2004; 23:386-94. [PMID: 14724567 DOI: 10.1038/sj.onc.1207071] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Survivin is a member of the inhibitor of apoptosis protein (IAP) family, which has been implicated in inhibition of apoptosis and control of mitotic progression. The finding that survivin is overexpressed in most human tumors but absent in normal adult tissues has led to the proposal of survivin as a promising therapeutic target for anticancer therapies. We decided to evaluate the effects of a ribozyme-based strategy for survivin inhibition in androgen-independent human prostate cancer cells. We constructed a Moloney-based retroviral vector expressing a ribozyme targeting the 3' end of the CUA(110) triplet in survivin mRNA, encoded as a chimeric RNA within adenoviral VA1 RNA. Polyclonal cell populations obtained by infection with the retroviral vector of two androgen-independent human prostate cancer cell lines (DU145 and PC-3) were selected for the study. Ribozyme-expressing prostate cancer cells were characterized by a significant reduction of survivin expression compared to parental cells transduced with a control ribozyme; the cells became polyploid, underwent caspase-9-dependent apoptosis and showed an altered pattern of gene expression, as detected by oligonucleotide array analysis. Survivin inhibition also increased the susceptibility of prostate cancer cells to cisplatin-induced apoptosis and prevented tumor formation when cells were xenografted in athymic nude mice. These findings suggest that manipulation of the antiapoptotic survivin pathway may provide a novel approach for the treatment of androgen-independent prostate cancer.
Collapse
Affiliation(s)
- Marzia Pennati
- Dipartimento di Oncologia Sperimentale, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Alain T, Thirukkumaran C, Morris DG, Urbanski SJ, Janowska-Wieczorek A, Lee PWK, Kossakowska AE. Lymphomas and Oncolytic Virus Therapy. ACTA ACUST UNITED AC 2003; 4:104-11. [PMID: 14556682 DOI: 10.3816/clm.2003.n.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There are several well-documented cases in medical literature of the remission of leukemias and malignant lymphomas following natural human viral infections. In the hope of being able to reproduce these spontaneous tumor regressions, investigators have studied various viruses with distinct oncolytic properties. The first attempts to treat patients with oncolytic viruses took place > 80 years ago; however, it achieved little success. With modern technologies and current knowledge of viruses and cancer, there is an expectation for the discovery of efficient oncolytic viral therapies. This article will review the current knowledge of oncolytic viruses in relation to the treatment of lymphoma.
Collapse
Affiliation(s)
- Tommy Alain
- Cancer Biology Research Group, University of Calgary, Alberta, Canada.
| | | | | | | | | | | | | |
Collapse
|
11
|
Salesse S, Verfaillie CM. BCR/ABL: from molecular mechanisms of leukemia induction to treatment of chronic myelogenous leukemia. Oncogene 2002; 21:8547-59. [PMID: 12476301 DOI: 10.1038/sj.onc.1206082] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
MESH Headings
- Antineoplastic Agents/therapeutic use
- Fusion Proteins, bcr-abl
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Stephanie Salesse
- Stem Cell Institute, Cancer Center and Department of Medicine, University of Minnesota Cancer Center, Minneapolis, Minnesota, MN 55455, USA
| | | |
Collapse
|