Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Boro Dropulic is active.

Publication


Featured researches published by Boro Dropulic.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Gene transfer in humans using a conditionally replicating lentiviral vector

Bruce L. Levine; Laurent Humeau; Jean D. Boyer; Rob-Roy MacGregor; Tessio Rebello; Xiaobin Lu; Gwendolyn K. Binder; Vladimir Slepushkin; Franck Lemiale; John R. Mascola; Frederic D. Bushman; Boro Dropulic; Carl H. June

We report findings from a clinical evaluation of lentiviral vectors in a phase I open-label nonrandomized clinical trial for HIV. This trial evaluated the safety of a conditionally replicating HIV-1-derived vector expressing an antisense gene against the HIV envelope. Five subjects with chronic HIV infection who had failed to respond to at least two antiviral regimens were enrolled. A single i.v. infusion of gene-modified autologous CD4 T cells was well tolerated in all patients. Viral loads were stable, and one subject exhibited a sustained decrease in viral load. CD4 counts remained steady or increased in four subjects, and sustained gene transfer was observed. Self-limiting mobilization of the vector was observed in four of five patients. There is no evidence for insertional mutagenesis after 21–36 months of observation. Immune function improved in four subjects. Lentiviral vectors appear promising for gene transfer to humans.


Journal of Virology | 2004

Antisense-Mediated Inhibition of Human Immunodeficiency Virus (HIV) Replication by Use of an HIV Type 1-Based Vector Results in Severely Attenuated Mutants Incapable of Developing Resistance

Xiaobin Lu; Qiao Yu; Gwendolyn K. Binder; Ziping Chen; Tatiana Slepushkina; John J. Rossi; Boro Dropulic

ABSTRACT We have constructed a human immunodeficiency virus type 1 (HIV-1)-based lentiviral vector expressing a 937-base antisense sequence against the HIV-1 envelope gene. Transduction of CD4+ T lymphocytes with this vector results in expression of the therapeutic antisense sequence and subsequent inhibition of productive HIV-1 replication. In this report, we examined the effect of antisense-mediated suppression on the potential development of virus escape mutants using a permissive T-cell line cultured under conditions that over serial passages specifically allowed for generation and amplification of mutants selected for by antisense pressure. In the resulting virus clones, we found a significant increase in the number of deletions at the envelope target region (91% compared to 27.5% in wild-type HIV). Deletions were most often greater than 1 kb in length. These data demonstrate for the first time that during antisense-mediated suppression of HIV, mutants develop as a direct result of selective pressure on the HIV genomic RNA. Interestingly, in clones where deletions were not observed, there was a high rate of A-G transitions in mutants at the antisense target region but not outside this region, which is consistent with those mutations that are predicted as a result of antisense-mediated modification of double-stranded RNA by the enzyme double-stranded RNA-specific adenosine deaminase. These clones were not found to be escape mutants, as their replicative ability was severely attenuated, and they did not replicate in the presence of vector.


Journal of Gene Medicine | 2005

Generation of a packaging cell line for prolonged large-scale production of high-titer HIV-1-based lentiviral vector.

Yajin Ni; Susan Sun; Ibe Oparaocha; Laurent Humeau; Brian M. Davis; Reuben Cohen; Gwendolyn K. Binder; Yung-Nien Chang; Vladimir Slepushkin; Boro Dropulic

An Erratum has been published for this article in Journal of Gene Medicine 7(6), 2005, 835.


Journal of Gene Medicine | 2004

Safe two-plasmid production for the first clinical lentivirus vector that achieves >99% transduction in primary cells using a one-step protocol.

Xiaobin Lu; Laurent Humeau; Vladimir Slepushkin; Gwendolyn K. Binder; Qiao Yu; Tatiana Slepushkina; Ziping Chen; Randall K. Merling; Brian M. Davis; Yung-Nien Chang; Boro Dropulic

We report the design of a unique two‐plasmid production system for the first lentiviral vector to be evaluated in humans, VRX496. VRX496 is an optimized VSV‐G pseudotyped vector derived from HIV‐1 that expresses antisense to the HIV envelope gene. We found that a two‐plasmid approach to production resulted in higher vector production titers when compared with a three‐plasmid approach, which is particularly important for vector production at the large scale. Therefore, we carefully designed a single packaging construct, VIRPAC, for safety by reducing its homology with VRX496 and by insertion of functionally validated genetic elements designed to reduce the risk of generation of a replication‐competent lentivirus (RCL). A native cis‐acting ribozyme is used to prevent read through into the envelope gene from the upstream gag‐pol genes in the packaging vector, thus preventing RNAs containing gag‐pol and env together for comparable safety to a three‐plasmid system. We demonstrate that there is no significant in vivo vector mobilization using a primary SCID‐hu mouse transplantation model, which correlates with the presence of an anti‐HIV payload and suggests that inclusion of antisense may be a useful tool to restrict mobilization in other vector constructs. Gene transfer is achieved using a one‐step transduction procedure that is simple and clinically translatable, which reaches stable transduction efficiencies of >99% in CD4+ T lymphocytes within 3 days of culture initiation. Copyright


Human Gene Therapy | 2011

Lentiviral vectors: their molecular design, safety, and use in laboratory and preclinical research.

Boro Dropulic

Lentiviral vectors have been successfully used in the clinic and they are increasingly being used for nonclinical applications. They are capable of stably transducing a broad range of mammalian cell types, including nondividing cells, with high efficiency. This review summarizes the evolving molecular design of lentiviral vectors, describing how they have improved since their first description. Lentiviral vector safety and issues surrounding genotoxicity are discussed. Examples of successful application of lentiviral vectors in laboratory and preclinical research are described. These include functional genomics, target validation, protein manufacturing, in vivo imaging, transgenic animals, and stem cell research.


PLOS ONE | 2010

Survival of the Fittest: Positive Selection of CD4+ T Cells Expressing a Membrane-Bound Fusion Inhibitor Following HIV-1 Infection

Janine Kimpel; Stephen E. Braun; Gang Qiu; Fay E. Wong; Michelle Conolle; Jörn E. Schmitz; Christian Brendel; Laurent Humeau; Boro Dropulic; John J. Rossi; Annemarie Berger; Dorothee von Laer; R. Paul Johnson

Although a variety of genetic strategies have been developed to inhibit HIV replication, few direct comparisons of the efficacy of these inhibitors have been carried out. Moreover, most studies have not examined whether genetic inhibitors are able to induce a survival advantage that results in an expansion of genetically-modified cells following HIV infection. We evaluated the efficacy of three leading genetic strategies to inhibit HIV replication: 1) an HIV-1 tat/rev-specific small hairpin (sh) RNA; 2) an RNA antisense gene specific for the HIV-1 envelope; and 3) a viral entry inhibitor, maC46. In stably transduced cell lines selected such that >95% of cells expressed the genetic inhibitor, the RNA antisense envelope and viral entry inhibitor maC46 provided the strongest inhibition of HIV-1 replication. However, when mixed populations of transduced and untransduced cells were challenged with HIV-1, the maC46 fusion inhibitor resulted in highly efficient positive selection of transduced cells, an effect that was evident even in mixed populations containing as few as 1% maC46-expressing cells. The selective advantage of the maC46 fusion inhibitor was also observed in HIV-1-infected cultures of primary T lymphocytes as well as in HIV-1-infected humanized mice. These results demonstrate robust inhibition of HIV replication with the fusion inhibitor maC46 and the antisense Env inhibitor, and importantly, a survival advantage of cells expressing the maC46 fusion inhibitor both in vitro and in vivo. Evaluation of the ability of genetic inhibitors of HIV-1 replication to confer a survival advantage on genetically-modified cells provides unique information not provided by standard techniques that may be important in the in vivo efficacy of these genes.


Molecular Therapy | 2011

Lentiviral Vector Platform for Production of Bioengineered Recombinant Coagulation Factor VIII

H. Trent Spencer; Gabriela Denning; Richard E. Gautney; Boro Dropulic; Andre Roy; Lajos Baranyi; Bagirath Gangadharan; Ernest T. Parker; Pete Lollar; Christopher B. Doering

Patients with hemophilia A present with spontaneous and sometimes life-threatening bleeding episodes that are treated using blood coagulation factor VIII (fVIII) replacement products. Although effective, these products have limited availability worldwide due to supply limitations and product costs, which stem largely from manufacturing complexity. Current mammalian cell culture manufacturing systems yield around 100xa0µg/l of recombinant fVIII, with a per cell production rate of 0.05xa0pg/cell/day, representing 10,000-fold lesser production than is achieved for other similar-sized recombinant proteins (e.g. monoclonal antibodies). Expression of human fVIII is rate limited by inefficient transport through the cellular secretory pathway. Recently, we discovered that the orthologous porcine fVIII possesses two distinct sequence elements that enhance secretory transport efficiency. Herein, we describe the development of a bioengineered fVIII product using a novel lentiviral-driven recombinant protein manufacturing platform. The combined implementation of these technologies yielded production cell lines that biosynthesize in excess of 2.5xa0mg/l of recombinant fVIII at the rate of 9xa0pg/cell/day, which is the highest level of recombinant fVIII production reported to date, thereby validating the utility of both technologies.


Journal for ImmunoTherapy of Cancer | 2017

A tandem CD19/CD20 CAR lentiviral vector drives on-target and off-target antigen modulation in leukemia cell lines

Dina Schneider; Ying Xiong; Darong Wu; Volker Nӧlle; Sarah Schmitz; Waleed Haso; Andrew Kaiser; Boro Dropulic; Rimas J. Orentas

BackgroundClinical success with chimeric antigen receptor (CAR)- based immunotherapy for leukemia has been accompanied by the associated finding that antigen-escape variants of the disease are responsible for relapse. To target hematologic malignancies with a chimeric antigen receptor (CAR) that targets two antigens with a single vector, and thus potentially lessen the chance of leukemic escape mutations, a tandem-CAR approach was investigated.MethodsAntigen binding domains from the FMC63 (anti-CD19) and Leu16 (anti-CD20) antibodies were linked in differing configurations to transmembrane and T cell signaling domains to create tandem-CARs. Expression on the surface of primary human T cells was induced by transduction with a single lentiviral vector (LV) encoding the tandem-CAR. Tandem-CARs were compared to single antigen targeting CARs in vitro and in vivo, and to an admixture of transduced cells expressing each CAR in vivo in immunodeficient (NSG) disease-bearing mice.ResultsTandem constructs efficient killed the Raji leukemia cell line both in vitro and in vivo. Tandem CARs generated less cytokine than the CD20 CAR, but similar to CD19 CARs, on their own. In co-culture experiments at low effector to target ratios with both single- and tandem- CAR-T cells, a rapid down-modulation of full-length CD19 expression was seen on leukemia targets. There also was a partial down-modulation of CD22, and to a lesser degree, of CD20. Our data also highlight the extreme sensitivity of the NALM-6 cell line to general lymphocyte-mediated cytotoxicity. While single and tandem constructs were effective in vivo in a standard setting, in a high-disease burden setting, the tandem CAR proved both effective and less toxic than an admixture of transduced T cell populations expressing single CARs.ConclusionTandem CARs are equally effective in standard disease models to single antigen specificity CARs, and may be both more effective and less toxic in a higher disease burden setting. This may be due to optimized cell killing with more moderate cytokine production. The rapid co-modulation of CD19, CD20, and CD22 may account for the ability to rapidly evolve escape mutants by selecting for leukemic clones that not require these target antigens for continued expansion.


Human Gene Therapy Methods | 2013

Rapid Generation of Stable Cell Lines Expressing High Levels of Erythropoietin, Factor VIII, and an Antihuman CD20 Antibody Using Lentiviral Vectors

Lajos Baranyi; Christopher B. Doering; Gabriella Denning; Richard E. Gautney; Kyle T. Harris; H. Trent Spencer; Andre Roy; Hatem Zayed; Boro Dropulic

Lentiviral vectors (LVs) are widely recognized as the most efficient method for the stable delivery of nucleic acid sequences into mammalian cells. Using erythropoietin (EPO), recombinant factor VIII (fVIII), and an anti-CD20 antibody as model proteins, we demonstrate advantages of LV-based gene delivery to achieve high production levels by transduced cells. Highly productive cell clones were able to incorporate up to 100 vector copies per cellular genome, without selection or gene amplification, and were isolated without extensive screening of a large number of clones. The LV transgenes were shown to be distributed throughout the genome, as visualized by fluorescent in situ hybridization. High-expressing clones producing 100-200u2009pg/cell/day of EPO were isolated and characterized. EPO production was demonstrated for at least 5½ months of continuous culture without selection, during which all the clones displayed high levels of glycosylation despite production levels at 10-20u2009g/liter. To demonstrate the utility of LV technology for multiple classes of proteins, cell lines producing fVIII and an anti-CD20 antibody were also developed. Cell clones demonstrating high levels of fVIII (100 clot units/ml and anti-CD20 antibody as high as 40-100u2009pg/cell/day) were isolated and characterized. LV-transduced cells and plasmid-transfected cells were compared for protein production per transgene copy. LV-transduced cells produced significantly higher levels of protein per copy of transgene than plasmid-transfected cells did. This study demonstrates the utility of LV technology for rapid generation of highly productive and stable cell lines over conventional plasmid transfection methods, significantly decreasing the time, cost, and risk of the manufacture of proteins and other complex biological molecules.


Neurosurgery | 2016

141 Phase I Trial of Genetically Modified Hematopoietic Progenitor Cells Facilitate Bone Marrow Chemoprotection and Enabling TMZ/O6BG Dose Escalation Resulting in Improved Survival.

Andrew E. Sloan; Hua Fung; Jane S. Reese; Lisa R. Rogers; Christopher Murphay; Hillard Lazrus; Boro Dropulic; Stan L. Gerson

INTRODUCTIONnGlioblastoma (GBM) is the most common malignant brain tumor with a median survival of 15 months despite surgery and aggressive radiochemotherapy. The most important mechanism of temozolomide (TMZ) resistance is the O-methylguanine-DNA methyltransferase (MGMT) gene that repairs temozolomide-induced DNA methylation. The MGMT inhibitor O-benzylguanine (BG) has demonstrated efficacy in depleting MGMT and maximizing tumor response in early-phase clinical trials. However, because MGMT expression is also low in hematopoietic cells, this has resulted in unacceptable bone marrow toxicity, and this approach has been abandoned. We hypothesized that chemoprotection of hematopoietic progenitor cells (HPC) with an MGMT mutant (MGMT-P140K) characterized by normal methyltransferase activity coupled with low affinity for BG, would maximize antitumor response while enabling patients to tolerate TMZ and BG dose escalation with minimal toxicity. We thus performed a phase I trial to test this hypothesis.nnnMETHODSnWe treated 10 consecutive newly diagnosed GBM patients with standard surgery and radiation, followed by transplantation with autologous CD34 hematopoietic progenitor cells engineered to express MGMT-P140K using a lentiviral vector using 3 different arms. To assess chemoprotection, patients blood counts and transgene marking were monitored during the treatment as was tumor growth and survival.nnnRESULTSnThe viral transduction rates were 2.5% to 75% and were clearly improved in the third arm with intrapatient dose escalation. P140K-MGMT gene markings in peripheral blood and bone marrow cells increased 3- to 26-fold with only mild (Grade 2-3) myelosuppression consistent with chemoselection and chemoprotection as hypothesized. Survival ranged from 20 to 36 months, which exceeded their recursive partitioning analysis-predicted survival by 1.9- to 3.2-fold suggesting clinical benefit (mean 2.0). Viral insertion site analysis failed to demonstrate clonal dominance.nnnCONCLUSIONnThese preliminary results demonstrate that this chemoprotection strategy is tolerable, safe, and facilitates TMZ and BG dose escalation resulting in increased survival. A phase II study is ongoing.

Collaboration


Dive into the Boro Dropulic's collaboration.

Top Co-Authors

Avatar

Laurent Humeau

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brian M. Davis

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carl H. June

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rimas J. Orentas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yajin Ni

Johns Hopkins University School of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge