Comprehensive and Unbiased Integration Site Analysis in Clinical Gene Therapy.

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2351-2351
Author(s):  
Richard Gabriel ◽  
Anna Paruzynski ◽  
Ralph Eckenberg ◽  
Cynthia C. Bartholomae ◽  
Ali Nowrouzi ◽  
...  

Abstract Retroviral vectors are commonly used gene delivery tools in clinical gene therapy providing stable integration and continuous gene expression of the transgene in the treated host cell. However, integration of the reverse transcribed vector DNA into the host genome is, by itself, a mutagenic eventthat may directly contribute to severe adverse events. The latter has dramatically been obbserved in individual cases in several, otherwise successful, gene therapy trials. Thus, a comprehensive analysis of the existing integration site pool in a transduced sample is indispensable to identify potential in vivo selection of affected cell clones and uncontrolled vector-induced cell proliferation. To date, there are several methods available to study the integration site distribution of retroviral vectors or other integrating elements as transposons. Each of these techniques makes use of restriction enzymes to digest the genomic DNA. To reveal particular vector integrations, a recognition motif of the used restriction enzyme has to be located in an appropriate distance to the integration locus in the host genome. Therefore, the genomic distribution of the recognition sequences directly impact the outcome of restriction enzyme dependent integration site analysis. We here report a validated genomic accessibility model which precisely determines the fraction of the human genome that can be analyzed with one reaction set up (i.e. restriction enzyme used). For our modeling, we used the clinically relevant linear amplification mediated PCR (LAM-PCR) as integration site analysis method of choice and the commonly used frequently cutting restriction enzymes (‘four-cutters’). We show that the most frequent four cutter motif (AATT) gives access to 54.5% of all possible integrations in the human genome, whereas the rarest distributed motif (CGCG) only identifies 2.9%. This restriction bias can be minimized by analyzing the same sample with different enzymes. A combination of the 5 most potent four cutter restriction enzymes gives access to 88.7% of the analyzable genome. Furthermore, we established an unbiased, non-restrictive integration site analysis technique based on (nr) LAM-PCR. Direct ligation of a single-stranded DNA sequence to the linear PCR product evades the need for restriction enzymes to recover integration sites. While standard LAM-PCR was done repeatedly with 3 different enzymes to detect integration sites present in lentivirally transduced single cell clones, nrLAM-PCR detected all integrations in these clones in one single reaction setup. This newly developed method comprehensively recovers genomic locations of integrating elements regardless of a restriction enzyme introduced bias. Our data show that the recovery rate of integration sites present in a transduced sample strongly depends on the restriction enzyme(s) used. However, we demonstrate that the genomic accessibility of viral integration sites indeed can be determined and minimized a priori, and that a non restrictive LAM-PCR approach circumvents the existing limitations. Analysis of the clonal inventory by these methods will allow determining the pharmacodynamics of insertional vectors with unprecedented precision, facilitating development and clinical testing of insertional vector systems.

Blood ◽  
2010 ◽  
Vol 115 (13) ◽  
pp. 2610-2618 ◽  
Author(s):  
Alice Gerrits ◽  
Brad Dykstra ◽  
Olga J. Kalmykowa ◽  
Karin Klauke ◽  
Evgenia Verovskaya ◽  
...  

Abstract Clonal analysis is important for many areas of hematopoietic stem cell research, including in vitro cell expansion, gene therapy, and cancer progression and treatment. A common approach to measure clonality of retrovirally transduced cells is to perform integration site analysis using Southern blotting or polymerase chain reaction–based methods. Although these methods are useful in principle, they generally provide a low-resolution, biased, and incomplete assessment of clonality. To overcome those limitations, we labeled retroviral vectors with random sequence tags or “barcodes.” On integration, each vector introduces a unique, identifiable, and heritable mark into the host cell genome, allowing the clonal progeny of each cell to be tracked over time. By coupling the barcoding method to a sequencing-based detection system, we could identify major and minor clones in 2 distinct cell culture systems in vitro and in a long-term transplantation setting. In addition, we demonstrate how clonal analysis can be complemented with transgene expression and integration site analysis. This cellular barcoding tool permits a simple, sensitive assessment of clonality and holds great promise for future gene therapy protocols in humans, and any other applications when clonal tracking is important.


2020 ◽  
Vol 117 (10) ◽  
pp. 5442-5452 ◽  
Author(s):  
Wenliang Wang ◽  
Maria Fasolino ◽  
Benjamin Cattau ◽  
Naomi Goldman ◽  
Weimin Kong ◽  
...  

Chimeric antigen receptor (CAR)-T immunotherapy has yielded impressive results in several B cell malignancies, establishing itself as a powerful means to redirect the natural properties of T lymphocytes. In this strategy, the T cell genome is modified by the integration of lentiviral vectors encoding CAR that direct tumor cell killing. However, this therapeutic approach is often limited by the extent of CAR-T cell expansion in vivo. A major outstanding question is whether or not CAR-T integration itself enhances the proliferative competence of individual T cells by rewiring their regulatory landscape. To address this question, it is critical to define the identity of an individual CAR-T cell and simultaneously chart where the CAR-T vector integrates into the genome. Here, we report the development of a method called EpiVIA (https://github.com/VahediLab/epiVIA) for the joint profiling of the chromatin accessibility and lentiviral integration site analysis at the population and single-cell levels. We validate our technique in clonal cells with previously defined integration sites and further demonstrate the ability to measure lentiviral integration sites and chromatin accessibility of host and viral genomes at the single-cell resolution in CAR-T cells. We anticipate that EpiVIA will enable the single-cell deconstruction of gene regulation during CAR-T therapy, leading to the discovery of cellular factors associated with durable treatment.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 2518-2518
Author(s):  
Kevin G Haworth ◽  
Lauren E Schefter ◽  
Zachary K. Norgaard ◽  
Jennifer E Adair ◽  
Hans-Peter Kiem

Abstract BACKGROUND A key event in the lifecycle of Human Immunodeficiency Virus (HIV) is permanent integration into the infected cells genome. In addition to allowing long-term persistence of the virus, this results in a trackable mark carried in all infected cells. Active HIV replication represses cellular pathways, preventing further cell division. This would imply that any specific integration site (IS) which is clonally expanded either during active or repressed viral infection arises from either a dormant/inactive virus, or is perturbing local gene expression, leading to increased cell proliferation. Alternatively, a cell carrying HIV provirus could proliferate due to T-cell specific antigen stimulation. By analyzing the patterns of integration sites detected in cell cultures and tissue samples from animal models of HIV infection, we can better understand the basic virology of integration site selection and determine what may potentially drive infected cells to persist despite effective treatment regimens. METHODS Jurkat reporter cell lines or primary human CD4+ cells were cultured and infected with various strains of HIV including both CCR5 and CXCR4 tropic viruses. Infected cells were cultured up to 21 days post infection, then analyzed for HIV proviral integration sites by next-generation sequencing. For in vivo studies, NSG mice were infused with human CD34+ hematopoietic stem/progenitor cells, resulting in a reconstituted human immune system including high levels of CD4+ T cells capable of sustaining HIV infection. After 16 weeks post-challenge, tissues were collected and subjected to integration site analysis for HIV proviral DNA. Identified integration sites were mapped and compared across multiple parameters to identify chromosomal regions and associated genes enriched for integration events, as well as clonally expanded cells in vivo. RESULTS Genome-wide analysis of HIV integration sites reveals a remarkably similar chromosomal landscape both in tissue culture infection of Jurkat cells and in vivo infection data (Figure 1), as well as across multiple HIV strains. As previously observed, the majority of integrations occur near or within gene coding regions thought to be actively transcribed at time of infection. However, certain areas of the genome, and even unique genes, are enriched for IS in individual samples. In addition to these genomic regions of enrichment, we also observe specific clonal outgrowth of unique integration events in genes previously unidentified in the literature. Three genes in particular exhibit a significant increase of integration events during acute infection which are 3x higher than predicted by random chance alone. We also observe integration events in genes that have been documented by other labs in HIV+ clinical patient samples, however in our active infection models, we do not see those specific genes enriched or expanded. This could indicate that these genes play a role in persistence that is only present during anti-retroviral therapy which suppresses active replication. CONCLUSIONS We have cataloged the most extensive HIV IS library to date in both relevant tissue culture models and in vivo infection studies, including over 245,000 unique integration events and three different HIV strains commonly used in research. Genome-wide correlation studies reveal regions significantly enriched for HIV integrations and genes which repeatedly exhibit clonal outgrowth in multiple animals. These types of studies are now being applied to human patient samples to determine if latency and persistence of infection can be mapped to unique integration events or genes of interest. Such information may indicate when and how the latent HIV reservoir is seeded and what types of therapy or treatments are most effective at targeting and eliminating these populations. Circos plot comparing HIV integrations sites (IS) identified either during in vitro cell culture infections (black bars), or in vivo infection studies using humanized mice (red bars). The outer ring is composed of human chromosomes each of which are divided into 25kB fragment bins. Total number of unique integration sites identified in each bin is represented by the height of the histogram bars. The in vitro IS concentric ring scale represents increments of 25 outwards up to 250 while the in vivo IS scales inwards in increments of 2 up to 16. Figure 1 Comparison of in vitro vs in vivo HIV Integration Sites. Figure 1. Comparison of in vitro vs in vivo HIV Integration Sites. Disclosures Adair: Rocket Pharmaceuticals: Consultancy, Equity Ownership.


Blood ◽  
2009 ◽  
Vol 114 (17) ◽  
pp. 3546-3556 ◽  
Author(s):  
Barbara Cassani ◽  
Eugenio Montini ◽  
Giulietta Maruggi ◽  
Alessandro Ambrosi ◽  
Massimiliano Mirolo ◽  
...  

Abstract Gene transfer into hematopoietic stem cells by γ-retroviral vectors (RVs) is an effective treatment for inherited blood disorders, although potentially limited by the risk of insertional mutagenesis. We evaluated the genomic impact of RV integration in T lymphocytes from adenosine deaminase-deficient severe combined immunodeficiency (ADA-SCID) patients 10 to 30 months after infusion of autologous, genetically corrected CD34+ cells. Expression profiling on ex vivo T-cell bulk population revealed no difference with respect to healthy controls. To assess the effect of vector integration on gene expression at the single-cell level, primary T-cell clones were isolated from 2 patients. T-cell clones harbored either 1 (89.8%) or 2 (10.2%) vector copies per cell and displayed partial to full correction of ADA expression, purine metabolism, and T-cell receptor-driven functions. Analysis of RV integration sites indicated a high diversity in T-cell origin, consistently with the polyclonal T-cell receptor-Vβ repertoire. Quantitative transcript analysis of 120 genes within a 200-kb window around RV integration sites showed modest (2.8- to 5.2-fold) dysregulation of 5.8% genes in 18.6% of the T-cell clones compared with controls. Nonetheless, affected clones maintained a stable phenotype and normal in vitro functions. These results confirm that RV-mediated gene transfer for ADA-SCID is safe, and provide crucial information for the development of future gene therapy protocols. The trials described herein have been registered at http://www.clinicaltrials.gov as #NCT00598481 and #NCT00599781.


2007 ◽  
Vol 82 (5) ◽  
pp. 2448-2455 ◽  
Author(s):  
S. Bleier ◽  
P. Maier ◽  
H. Allgayer ◽  
F. Wenz ◽  
W. J. Zeller ◽  
...  

ABSTRACT Analysis of the fate of retrovirally transduced cells after transplantation is often hampered by the scarcity of available DNA. We evaluated a promising method for whole-genome amplification, called multiple displacement amplification (MDA), with respect to even and accurate representation of retrovirally transduced genomic DNA. We proved that MDA is a suitable method to subsequently quantify engraftment efficiencies by quantitative real-time PCR by analyzing retrovirally transduced DNA in a background of untransduced DNA and retroviral integrations found in primary material from a retroviral transplantation model. The portion of these retroviral integrations in the amplified samples was 1.02-fold (range 0.2, to 2.1-fold) the portion determined in the original genomic DNA. Integration site analysis by ligation-mediated PCR (LM-PCR) is essential for the detection of retroviral integrations. The combination of MDA and LM-PCR showed an increase in the sensitivity of integration site analysis, as a specific integration site could be detected in a background of untransduced DNA, while the transduced DNA made up only 0.001%. These results show for the first time that MDA enables large-scale sensitive detection and reliable quantification of retrovirally transduced human genomic DNA and therefore facilitates follow-up analysis in gene therapy studies even from the smallest amounts of starting material.


2019 ◽  
Author(s):  
Ping Zhang ◽  
Devika Ganesamoorthy ◽  
Son Hoang Nguyen ◽  
Raymond Au ◽  
Lachlan J. Coin ◽  
...  

AbstractVector integration site analysis can be important in the follow-up of patients who received gene-modified cells, but current platforms based on next-generation sequencing are expensive and relatively inaccessible. We analyzed polyclonal T cells transduced by a gammaretroviral vector, SFG.iCasp9.2A.ΔCD19, from a clinical trial. Following restriction enzyme digestion, the unknown flanking genomic sequences were amplified by inverse polymerase chain reaction (PCR) or cassette ligation PCR. Nanopore sequencing could identify thousands of unique integration sites within polyclonal samples, with cassette ligation PCR showing less bias. The assay is scalable and requires minimum capital, which together enable cost-effective and timely analysis.


Sign in / Sign up

Export Citation Format

Share Document