期刊论文详细信息
Virology Journal
Serotype-dependent recombinant adeno-associated vector (AAV) infection of Epstein–Barr virus-positive B-cells, towards recombinant AAV-based therapy of focal EBV + lymphoproliferative disorders
Mazyar Ziyaeyan1  Santosh Kumar Guru2  Mehrdad Ravanshad3  Elham Ahmadi4  Sandeep S. Jubbal5  Joyce Fingeroth5  Rajesh Panigrahi5  Gao Guangping6  Jun Xie6 
[1] Alborzi Clinical Microbiology Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran;Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, 01605, Worcester, MA, USA;Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14155-331, Tehran, Iran;Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14155-331, Tehran, Iran;Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, 01605, Worcester, MA, USA;Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Medical School, 364 Plantation Street, 01605, Worcester, MA, USA;Horae Gene Therapy Center, University of Massachusetts Medical School, 364 Plantation Street, 01605, Worcester, MA, USA;
关键词: rAAV;    EBV;    B-cells;    Focal tumors;    PTLD;    Mononucleosis;    Suicide gene;    Transfection;   
DOI  :  10.1186/s12985-021-01695-w
来源: Springer
PDF
【 摘 要 】

BackgroundB-cell proliferative disorders, such as post-transplant lymphoproliferative disease (PTLD), are increased among persons afflicted by T-cell compromise. Most are Epstein–Barr virus (EBV) + and can first present with a focal lesion. Direct introduction of oncolytic viruses into localized tumors provides theoretical advantages over chemotherapy, immunotherapy and radiation therapy by reducing systemic toxicity. Despite extensive study as a vehicle for gene therapy, adeno-associated viruses (AAV) have rarely been applied to human cancer research due to technical and theoretical obstacles. Moreover, human B-cells have historically been described as resistant to AAV infection. Nonetheless, advances using different recombinant (r)AAV serotypes with unique tropisms to deliver cytotoxic therapy suggested a localized anti-tumor approach was feasible.MethodsAs a prelude to the development of a therapeutic vehicle, the ability of fifteen distinct EGFP-bearing rAAV serotypes to transduce human B-cells, including primary, immortalized, and B-cell tumor lines ± EBV was assessed by confocal microscopy, flow cytometry and subsequently cell viability assay.ResultsRank order analysis revealed augmented transduction by rAAV6.2 and closely related virions. EBV infection of EBV-negative B-cell tumor lines and EBV immortalization of primary B-cells increased susceptibility to rAAV6.2 transduction. As a proof of concept, transduction by rAAV6.2 encoding herpes simplex virus type 1 (HSV1)-thymidine kinase (TK) eliminated TK-negative rhabdomyosarcoma cells and diminished viability of transduced B-cell lines upon incubation with ganciclovir.ConclusionsrAAV serotypes differentially transduce human B-cell lines reversing the dogma that human B-cells are refractory to AAV infection. EBV + B-cells display increased susceptibility to rAAV6.2 infection, uncovering a new method for improved nucleic acid transfer into transfection-resistant B-cell lines. The introduction of a functional suicide gene into the rAAV6.2 genome identifies a candidate vector for the development of rAAV-based oncolytic therapy targeting focal EBV-bearing B-lymphoproliferative disorders.

【 授权许可】

CC BY   

【 预 览 】
附件列表
Files Size Format View
RO202112041901452ZK.pdf 3082KB PDF download
  文献评价指标  
  下载次数:5次 浏览次数:6次