Synopsis
Current hematopoietic stem cell (HSC) gene therapy approaches rely on the collection of patient HSCs, followed by their culture and expansion in vitro, their modification using γ-retrovirus or lentiviral vectors, and their re-infusion into myelo-conditioned patients. While this approach has been successfully used in numerous clinical trials, its reliance on the extended ex vivo culture of patient HSCs comes with a set of disadvantages. Culturing HSCs in the presence of a cytokine cocktail to facilitate their expansion and transduction is thought to negatively impact the long-term viability of HSCs, and their homing and repopulation capacity. Importantly, the requirement for myeloablative regimens in patients represents a critical risk factor and results in considerable morbidity. In addition to these biological problems, the process of ex vivo HSC manipulation is also challenging from a logistics and regulatory standpoint, as manipulations must be performed in specialized, accredited centers. These complexities lead to a high price of such gene therapeutic regimens, and subsequently severely limited patient access to these treatments. In vivo HSC gene therapy approaches aim to simplify the gene therapy process by eliminating the need for ex vivo handling of patient HSCs. The in vivo approach can be subdivided into three methods. One method is based on the direct modification of HSCs in the bone marrow following intraosseous injection. Another involves intravenous injection of gene delivery vectors that home to HSCs in bone marrow. We have developed an alternative approach in which HSCs from the bone marrow are mobilized into peripheral blood, become gene modified upon intravenous vector administration, and then re-engraft in the bone marrow. We will provide examples of these three approaches and then discuss the advantages and disadvantages of in vivo HSC gene therapy.
Keywords: intravenous, intraosseal, viral vectors, mobilization
Introduction
HSCs in the bone marrow
The majority of HSCs reside in so-called stem cell niches regions of the bone marrow in which non-hematopoietic cells interact with HSCs and regulate their dormancy and self-renewal or their differentiation and expansion 1. However, a small fraction of HSCs circulate in the peripheral blood under steady state conditions 2. HSCs that leave the bone marrow under steady state conditions provide a means of exchange between different stem cell niches as well as a way to react to local damage within hematopoietic tissues. Even under normal conditions, circulating HSCs appear to be able to rapidly re-engraft in bone marrow so that there is a cycle of constant egress and re-engraftment of HSCs 3. HSC regulation and retention within the bone marrow stem cell niche is mediated through multiple interactions between HSC surface receptors and their respective ligands expressed or secreted by surrounding cells, including osteoblasts, sinusoidal endothelial and perivascular cells.
HSC mobilization
The induced egress of HSCs is referred to as stem cell mobilization. While egress of HSCs from the bone marrow can be observed in response to stress, for example following injury of hematopoietic organs, stimulated mobilization is commonly achieved through drug administration. The most commonly used mobilizing agent is recombinant human G-CSF, which is given in the form of subcutaneous injections for 5 days, and leads to efficient mobilization of both HSCs as well as more differentiated cells 2. Another class of mobilizing agents are CXCR4 antagonists, most prominently the FDA-approved drug Plerixafor/AMD3100. CXCR4 antagonists lead to more rapid mobilization of HSCs than G-CSF4, and are thought to cause mobilization solely through disruption of the SDF-1-CXCR4 axis. AMD3100 has been shown to synergize with G-CSF mobilization. Due to its higher mobilization power, the combination of G-CSF and AMD3100 is used as a regimen in poor HSC mobilizers such as chemotherapy patients 5. In a similar manner soluble SCF is able to interrupt the connection between membrane-bound SCF and c-kit, even though SCF is considered to be a slow mobilizing agent and has to be administered for several days 6. Another class of mobilizing agents target VLA-4, and both VLA-4 binding antibodies 7 and small molecules 8 are able to rapidly mobilize HSCs. VLA-4 binding agents are also thought to have a synergistic or additive effect when used together with G-CSF and AMD3100 9. One VLA-4 inhibitor, the small molecule BIO5192, was shown to efficiently and rapidly mobilize HSCs. Furthermore, BIO5192 can be combined with G-CSF alone or with AMD31000 to increase levels of mobilized HSCs. BOP (N-(benzenesulfonyl)-L-prolyl-L-O-(1-pyrrolidinylcarbonyl)tyrosine), a small molecule targeting α9β1/α4β1 integrins also rapidly mobilizes long-term multi-lineage reconstituting HSC 10. Synergistic engraftment augmentation is observed when BOP is co-administered with AMD3100.
Examples for in vivo HSC gene therapy (Fig. 1)
Fig. 1. Schematic representation of four approaches for in vivo HSC gene therapy.
The upper left panel shows an example of in vivo HSC gene editing using non-viral nanoparticles injected intravenously (IV) into mice. Site-specific triplex-forming peptide nucleic acids (PNA) interact site-specifically with genomic DNA via strand invasion resulting in the displacement of a DNA strand. This recruits endogenous DNA repair proteins to initiate site-specific modification of the genome when single-stranded donor DNAs are co-delivered. The upper right panel shows a study in dogs with a mutation causing SCID-X1. A foamy virus vector carrying the γC gene capable of phenotypic correction of the disease, is delivered intravenously. The lower left panel depicts an intraosseal (IO) approach of gene delivery into bone marrow HSCs. In this example, a lentivirus vector carrying a human FVIII clotting factor expression cassette is injected via a syringe pump into the femurs of mice. The lower right panel shows an in vivo HSC gene transfer approach that involves the mobilization of HSCs from the bone marrow by subcutaneous (SC) injections of G-CSF/AMD3100. While HSC are in the peripheral blood, a capsid-modified, integrating helper-dependent adenovirus vector (HDAd5/35++) is injected intravenously. HSC transduced in the periphery home back to the bone marrow where they persist long-term.
Example 1: Intravenous injection of triplex-forming peptide nucleic acids (PNAs)
PNAs are designed to bind site-specifically to genomic DNA via strand invasion and formation of PNA/DNA/PNA triplexes with a displaced DNA strand. PNAs consist of a charge-neutral peptide-like backbone and nucleobases enabling high affinity hybridization with DNA. PNA/DNA/PNA triplexes can be used to modify DNA by recruiting endogenous DNA repair proteins to initiate site-specific modification of the genome when single-stranded ‘donor DNAs’ are co-delivered as templates containing the desired sequence modification 11.
A recent study reported in vivo HSC gene editing in mice using intravenously injected triplex-forming PNAs in combination with SCF given i.p. 3 hours before the PNAs 12. Treatment of transgenic mice carrying a β-globin/GFP reporter transgene with PNAs and single-stranded donor DNA yielded gene editing in mouse CD117+ cells at frequencies up to 1% with a single treatment. In a thalassemic mouse model, in vivo treatment resulted in a gene editing frequency of almost 4% in total bone marrow mononuclear cells and 6.9% in HSCs, resulting in amelioration of the disease phenotype. The authors reported that, based on their ex vivo studies, SCF could enhance PNA-mediated gene editing in vivo. We speculate that SCF treatment might also have resulted in HSC mobilization and thus caused better gene transfer. A non-viral PNA-based platform for in vivo HSC gene editing has the advantage of circumventing host immune responses and, if it can be scaled-up, has great potential over designer nucleases-based approaches for gene editing that introduce an active nuclease into cells, which can lead to off-target cleavage in the genome and can affect the viability and function of HSCs.
Example 2: Intravenous injection of foamy virus vectors in dogs
Current VSVG-pseudotyped lentivirus vectors are not suitable for direct in vivo injection, since VSVG is rapidly inactivated by human complement 13. In this respect, several pseudotyping partners have emerged in the field, such as the complement-resistant gibbon ape leukemia virus (GaLV) or the endogenous feline leukemia virus RD114 glycoproteins. The Kiem group has used foamy virus (FV) vectors, which are non-pathogenic integrating retroviruses with properties that distinguish them from other viral vectors. Unlike VSV-G pseudotyped lentiviral vectors, FV vectors are resistant to human serum inactivation 14, which gives them a specific advantage in the context of in vivo delivery. FV vector integrations are less frequent near transcriptional start sites when compared to γ-retrovirus vectors, and FV vectors also do not show a preference for integrating within highly expressed genes, as has been observed for lentivirus vectors 15. Importantly, Hendrie et al. also showed that FV vector proviruses are less likely to activate nearby genes even if they do integrate close to them 16. A number of groups demonstrated that FV vectors are highly proficient in gene transfer to HSCs from both human and large animals, such that promising therapeutic constructs can be efficiently evaluated under preclinical settings. Safety of FV vectors for gene therapy applications has also been established in dogs 17 and in human SCID repopulating cells 18. FV vectors thus offer a unique combination of properties that makes them a potentially superior choice for retrovirus gene therapy and for therapeutic gene corrections with diseases such as SCID-X1. We have used the canine SCID-X1 model, which is a naturally occurring genetic disease caused by inactivating mutations in the IL2RG gene that encodes the shared common gamma chain (γC) signaling component of the IL-2, IL-4, IL-7, IL-9, and IL-15 receptors 19. Canine SCID-X1 provides an excellent pre-clinical model as it manifests nearly identical characteristics compared to human SCID-X1. Both disorders are characterized by absent thymic T-cell development leading to an absence of mature peripheral T-cells, resulting in the lack of T-cell mediated immune responses and dysregulated B cell germinal center responses that, in turn, lead to low IgA, IgM and IgG levels, absence of lymph nodes, failure to thrive, and early infant mortality due to viral and/or bacterial infection 20.
We and others have explored in vivo gene therapy 21. In collaboration with Dr. Peter Felsburg we have investigated in vivo delivery using FV vectors in SCID-X1 dogs. Intravenous injection of a FV vector expressing a codon-optimized human γC gene in newborn SCID-X1 dogs resulted in expansion of corrected CD3 T lymphocytes that expressed the CD4 or CD8 co-receptors, underwent antigen receptor gene rearrangement, and were functionally mature enough to respond to T cell mitogens. Furthermore, FV integration site analysis demonstrated that the provirus did not integrate near known proto-oncogenes, and there was no evidence of clonal expansion that indicated a leukemic transformation 21. We also evaluated generation of specific antibody responses and immunoglobulin class-switching in treated animals after immunization with the T cell dependent neoantigen bacteriophage, phiX174. This neoantigen has been used to evaluate dogs and humans with SCID-X1 before and after bone marrow transplantation and gene therapy. We found that treated animals showed a primary and secondary antibody response that is very similar to that seen in normal human controls, indicating that our treatment restored both the B and T cell cytokine signaling that is required for class switching and memory responses to this neoantigen. Altogether, our results demonstrate that in vivo gene therapy using FV vectors is safe. In vivo delivery of γC expressing FV vector to dogs resulted in immune reconstitution with gene-corrected T-cells but the treated animals still developed infections and had low immunoglobulin levels and marking levels in granulocytes and monocytes were very low (0.6%), indicating that further increase in the efficacy of HSC transduction is required to achieve long-term phenotypic correction. There are several ways in which this type of in vivo gene therapy can be further optimized. The kinetics of immune reconstitution may be enhanced by modifying FV vector design, for example, by using a stronger promoter in place of the currently used human elongation factor-1 alpha promoter (EF1α) to drive expression of γC. In addition, gene marking in other cell lineages that do not have a selective advantage like T lymphocytes may be increased by more efficiently targeting HSCs. This could be accomplished by using mobilizing agents to increase the number of circulating HSCs in peripheral blood or by the in vivo selection of gene-modified HSCs. Importantly, the in vivo gene therapy procedure developed here for SCID-X1 is highly portable and could be disseminated worldwide for the treatment of additional genetic blood disorders.
Example 3: Intraosseal lentivirus injection in mice
Although lentiviral vectors (LV) have traditionally been used in ex vivo HSC gene therapy, recent advances showed that LV can have utility for in vivo gene therapy. As an example, we will discuss LV-mediated FVIII gene transfer by intra-femoral injection in mice. LV-mediated gene transfer into HSCs resulted in stable integration of FVIII gene into the host genome, leading to a persistent therapeutic effect 22–24. This IO infusion method was improved by using a syringe pump to slowly infuse LVs into the bone marrow so that more vectors can be retained for longer in the marrow cavity in order to achieve high levels of transduction of bone marrow cells 22. In another study, a single infusion of GFP-LV achieved persistent GFP expression in 10–50% of HSCs for up to 160 days, indicating that IO LV delivery mediates sustained transduction of hematopoietic stem/progenitor cells 22,25. Furthermore, IO delivery of E-F8-LVs encoding the FVIII gene driven by a ubiquitous EF1α promoter produced FVIII expression in various types of cells including antigen presenting cells, and high levels of FVIII were initially secreted into the circulation. However, a high-titer inhibitory antibody response to FVIII was rapidly induced, which completely eliminated functional FVIII activity. In contrast, a single IO infusion of G-F8-LVs driven by a megakaryocyte-specific promoter produced long-term stable expression of FVIII in platelets and initiated a long term corrected hemophilia phenotype 22. Most importantly, this strategy was also proven successful in hemophilia A mice with pre-existing inhibitory antibodies 22. This is because FVIII stored in α-granules of platelets is protected from high-titer anti-FVIII antibodies, and the locally excreted FVIII following platelet activation that participates directly in clot formation is functionally more potent than plasma FVIII.
Another example for IO injection was described by Frecha et al.13. They used LVs displaying stem cell factor and thrombopoietin, which allows for targeting of the vector particles to HSCs expressing c-Kit and c-Mpl. These vectors were more efficient in transducing CD34+ cells after intrafemural injection into mice with engrafted human CD34+ cells. However, the overall percentage of GFP+ cells in human CD45+ cells in the BM was only 0.7%.
While IO LV injection in mice is technically difficult and challenging to standardize due to limited space for the injected virus in the bone, IO injection could be more feasible in large animals and humans. This is supported by studies with IO transplantation of HSCs. Intraosseous delivery of donor hematopoietic cells has been tested to mitigate the inevitable early loss of cells given I.V. to high blood volume organs, like liver and lung. It was first attempted in mice using different approaches 26–28 and later in larger animals, canines 29, non-human primates 30 and cancer patients 31,32. The source of cells was bone marrow, cord blood, or mobilized peripheral blood and recipients were either myeloablated or received reduced intensity or no conditioning. Regarding the mouse, the overall assessment is that, despite multiple efforts, the outcome has not been better than the intravenous infusion, most likely because of anatomic constraints. In larger animals, the results differ depending on the number of cells used or the conditioning. The effort of using cord blood via the intraosseous route is in its early stages and many variables influencing the outcome need to be firmly ascertained; these include the optimum volume to be injected, the cell concentration, the number of injection sites and the optimum conditioning.
Example 4: HSC transduction using adeno-associated virus vectors
Although we are not aware of published data describing direct in vivo HSC transduction following intravenous adeno-associated virus (AAV) vector delivery, this vector system has potential for in vivo HSC gene addition and targeting. Furthermore, AAV has proved highly promising in both preclinical and clinical safety and efficacy studies for hepatocyte targeted gene therapy 33,34. Sveral ex vivo studies with AAV and HSCs demonstrate their potential utility for in vivo HSC transduction, although HSCs from some but not all species can be efficiently transduced by AAV when the best serotype and culture conditions are used. AAV1 vectors, for example, can transduce mouse Lin−Sca1+Kit+ (LSK) cells, whereas CD34+ cells from cynomolgus monkeys are poorly transduced by AAV vectors derived from serotypes 1–10 35. A number of groups have shown that human HSCs can be readily transduced with AAV6 based vectors 35,36, and vectors with AAV6 capsids containing surface exposed tyrosine to phenylalanine mutations transduce human HSCs with the highest efficiency 35,37.
The ability of AAV vectors to efficiently deliver their genomes to the nucleus of HSCs has led to the development of gene targeting strategies, as their single stranded vector genomes can be efficiently used as donor templates during the process of homologous recombination directed targeted integration 38. Two recent studies demonstrated that co-delivery of an AAV6 donor template in combination with a target site directed endonuclease leads to efficient ex vivo gene editing in human HSCs that retain the ability to engraft in NSG mice and differentiate into multiple hematopoietic lineages 39. In vivo engraftment studies have also successfully been performed in mice using self-complementary AAV1 (scAAV) vector transduced LSK cells. Unlike classical AAV vectors, scAAV vectors do not require second strand synthesis or intermolecular annealing of their genomes to initiate transgene expression, so typically provide more efficient gene marking. Both primary and secondary transplant recipients showed up to 7% transgene expression in peripheral blood 40. More recently gene edited HSCs that were engineered using ZFN endonucleases and AAV6-derived targeting vectors showed long-term engraftment and differentiation in NSG mice 41. A similar ZFN plus AAV6 based ex vivo approach to HSC gene editing has subsequently been used for the treatment of X-linked chronic granulomatous disease, with up to 11% of bone marrow cells in primary recipients containing an introduced copy of the gp91phox subunit of the NADPH oxidase 42.
AAV does have some potential disadvantages for direct in vivo use such as the relatively high cost of production and limited knowledge of the in vivo tropism for AAV serotypes that can transduce HSCs ex vivo. For example, AAV6 shows high-affinity for skeletal muscle upon IV administration 43, which will likely impede attempts to target HSCs in situ. Furthermore, while efficient gene expression in the liver can theoretically be achieved from episomal AAV vector genomes, HSC gene therapy requires vector integration, a process that is not well studied in HSCs.
Example 5: In vivo transduction of primitive HSCs after mobilization and intravenous injection of integrating helper-dependent adenovirus vectors
Human adenovirus (Ad) has long been used as a gene transfer vector to various tissues. However, early studies showed that the most commonly used serotype, Ad5, was incapable of efficiently transducing HSCs. In contrast, other less studied Ad serotypes, especially those belonging to human species B, showed much more promise in their ability to transduce HSCs. A number of species of B Ads, including serotypes 11, 16, 21, 34, 35, and 50 use CD46 as a receptor 44, a membrane protein expressed on all nucleated cells in humans. Its main function is to protect self-tissue from inadvertent killing by the complement system, but it also has important signaling functions, for example in the regulation of T-cell activity 45. We and others have found that CD46 is uniformly expressed on all CD34+ cells 44,46.
In order to harness the well-understood biology of Ad5, the tools to manipulate and vectorize Ad, as well as the enhanced HSC transduction capabilities of species B Ads, fiber-chimeric vectors containing species B fibers on an Ad5 capsid were developed. Vectors carrying the fiber of CD46-tropic adenovirus type 35 (Ad5/35) were subsequently shown to efficiently transduce human HSCs 46–50. Importantly, after intravenous injection into mice and non-human primates, first-generation Ad5/35 vectors did not cause liver toxicity 51–53, a problem seen with serotype 5 (Ad5) based vectors 54. Moreover, our group introduced mutations in the Ad35 fiber knob to further increase its affinity toward the Ad35 cellular receptor CD46, leading to increased transduction capabilities when targeting HSCs 55.
To further increase the suitability as a gene transfer vector for HSCs, helper-dependent adenovirus vectors (HDAds) have been developed 56–59. These vectors are devoid of all viral genes and only contain minimal amounts of viral DNA. This completely abolishes the leaky expression of adenoviral genes that had been shown to lead to cytotoxicity in HSCs that have been transduced with earlier Ad vectors that still encoded these genes 59,60.
The goal of HSC gene therapy is the lifelong correction of an underlying genetic disease. To fulfill this requirement integration of the therapeutic transgene is generally required so that gene-corrected HSCs are able to give rise to gene corrected progeny cells. Ad5 however, does not actively integrate its genetic material into the genome of infected cells. To counteract this shortcoming, HDAd vectors can be armed with transposon-based transgene integration systems. Multiple Class II DNA transposons display activity in human cells, including Tol2, piggyBac, and Sleeping Beauty (SB). Tol2 and piggyBac have the propensity to integrate transposons in and around actively transcribed genes 61. In our studies we have used a hyperactive Sleeping Beauty (SB100x) transposase system 62,63. The SB transposase, when co-expressed in trans from a second vector, recognizes specific DNA sequences (inverted repeats-“IRs”) flanking the transgene cassette and triggers its integration into TA dinucleotides of chromosomal DNA. Unlike retrovirus integration, SB-mediated integration does not depend on the transcriptional status of the targeted genes nor on cellular DNA repair proteins 64. Studies with human cell lines and mouse hepatocytes in vitro and in vivo showed that SB-mediated transgene integration is random and has not been associated with the activation of proto-oncogenes 63.
In our experience, intravenous injection of adenovirus vectors in mice and non-human primates does not lead to transduction of HSCs in the bone marrow, even if the vector is capable to efficiently transduce HSCs in vitro 51,52. We therefore decided to mobilize HSCs from the bone marrow and transduced them in the periphery with integrating HDAd5/35++ vectors.
In our studies, we employed the SB100x vector platform in the context of HSC gene therapy and used a human CD46 transgenic mouse model to study stable transfer a reporter gene cassette into HSCs in vivo 65. HSCs were mobilized from the bone marrow of mice into the peripheral blood through administration of G-CSF and AMD3100, after which integrating HDAd5/35 vectors were injected intravenously. We were able to show in vivo transduction following this regimen as reporter gene-positive LSK cells could be detected in the bone marrow of mice (Fig. 2A). We saw that 6.5% of bone marrow LSK cells expressed a reporter transgene at 20 weeks post in vivo transduction. Furthermore, HSCs transduced in this way could give rise to progenitor colonies consisting of reporter gene-positive progeny cells (Fig. 2B) and at 20 weeks after in vivo transduction 9% of CFU progenitor colonies were positive for the transgene. This showed that the reporter gene had been integrated into the HSC genome and that the gene modified cells were still functional stem cells capable of proliferation and differentiation. Importantly, CFU assays showed an increase in GFP-positive colonies over time indicating transduction of long-term surviving and potentially self-renewing cells capable of reconstitution of hematopoiesis in lethally irradiated transplant recipients. Moreover, we demonstrated that HSCs modified after in vivo transduction could rescue lethally irradiated mice upon transplantation and that the recipients of these transplants showed reporter gene expression in all hematopoietic lineages up to 16 weeks after transplantation. In addition, successful HSC transduction could be shown in a humanized mouse model when using a similar in vivo transduction regimen.
Fig. 2. GFP marking in bone marrow HSCs after in vivo transduction.
HSCs were mobilized in human CD46 transgenic mice by s.c. injections of human recombinant G-CSF (5μg/mouse/day, 4 days) followed by an s.c. injection of AMD3100 (5mg/kg) eighteen hours after the last G-CSF injection. Two integrating HD-AD5/35++ vectors at a total dose of 8×1010 viral particles were injected IV one hour after AMD3100. One Ad (HDAd-GFP) contains a GFP gene under the control of the EF1α promoter, and the transgene cassette is flanked by inverted repeats that are recognized by the Sleeping Beauty transposase. The second Ad vector (HDAd-SB) expresses the hyperactive Sleeping Beauty transposase SB100x in trans. Animals were sacrificed 4, 8, 12, or 20 weeks after Ad transduction and bone marrow cells were isolated (n≥=10). (A) Shown is the percentage of GFP-positive HSCs (lin−, Sca1+, cKit+ -“LSK”) present in the bone marrow. (B) GFP expression in colony forming units. Bone marrow cells were lineage depleted via MACS followed by collection of GFP-positive cells via FACS. Cells were then plated in CFU assays and colonies were scored 12 days after plating.
Data from Richter M, Saydaminova K, Yumul R, et al. In vivo transduction of primitive mobilized hematopoietic stem cells after intravenous injection of integrating adenovirus vectors. Blood 2016; 128(18):2206–2217.
In addition to the general advantages of in vivo HSC gene therapy, the use of adenoviral vectors for this application provides a number of additional advantages. HDAd vectors have a relatively large cloning capacity of up to 34 kb. However, it has to be noted that when using the SB transposase, efficient transposition is only observed for cassettes of up to 10 kb in size. Nevertheless, the adenoviral in vivo delivery approach could also be employed in the context of a targeted nuclease approach in which the large cloning capacity of Ad vectors could be exploited to deliver both a nuclease and a homologous recombination template for a gene of interest using the same vector.
Another major advantage of the use of adenoviral gene transfer vectors is their ease of production. HDAd vectors can be easily produced to high titers and scale up is usually uncomplicated since the production of these vectors does not rely on large-scale transfection of genetic material. These advantages also result in a relatively low vector production cost, often orders of magnitude lower than those encountered for lentiviral or AAV vectors. A further advantage of an Ad vector platform is the ability to lyophilize vector preparations to ensure long term stability even when stored at 4°C 66.
Discussion
In vivo HSC gene transfer achieved using a minimally invasive manner and without the need for stem cell harvest or transplantation should simplify HSC gene therapy. Furthermore, by avoiding ex vivo HSC manipulation, the risk of cell differentiation or loss of homing/engraftment capability is avoided, as there is no need for the extraction and purification of target cells. In vivo gene delivery also bypasses the issue concerning the markers that enable the isolation of true primitive HSCs. In vivo gene transfer could target all HSCs, including those that are missed by different purification criteria. Finally, in vivo HSC transduction should eliminate the need for myeloconditioning with chemotherapy drugs. Preconditioning results in considerable early morbidity owing to transitory blood-cell depletion, immunodeficiency and mucosal damage, which place the recipient at risk of severe infection 67. It also causes delayed morbidity owing to the risk of developing chemotherapy-induced secondary tumors and infertility. Several HSC gene-therapy trials have attempted to alleviate the morbidity associated with preconditioning by lowering the dosage and combination of chemotherapeutic drugs. However, the impact of changing these drug regimens on the risks and benefits of the therapy is yet to be determined in broader comparative studies and through long-term patient follow-up. Finally, the technical complexity and high cost of current ex vivo HSC gene therapy is a barrier to a widespread application for common diseases. As an example, Strimvelis™, a HSC gene therapy for ADA-SCID that has been approved for marketing in the European Union earlier this year, comes at a price of close to $700,000 per treatment and the regimen is only available at a single center in Europe making access to it difficult even within the developed countries of Europe
In vivo HSC gene transfer approaches do pose a number of potential problems, with the most important ones being due to direct contact between the gene transfer vector and the patient, which can trigger innate and adaptive immune responses. This problem is specifically pronounced for approaches involving intravenous vector injection. Viral vectors and non-viral nanoparticles are taken up by monocytes and macrophages in various tissues, specifically within the liver and spleen, and the incoming particle and DNA or RNA is sensed by innate immune mechanism. This results in release and activation of pro-inflammatory cytokines 68. It is thought that AAV vectors provoke weaker innate immune responses than other viral vectors such as Ad or LV 69, however, at this point no direct comparisons of these vector systems with regards to innate toxicity have been made following IV injection. In our studies with HDAd5/35++ vectors we found that the release of proinflammatory cytokines upon vector administration is increased by mobilization. Mobilization, specifically regimens that involve G-CSF, triggers acute leukocytosis so that more monocytes in the circulation can come into contact with the IV injected vector. Acute cytokine release however can be prevented by pre-treatment with glucocorticoids 70, or anti-IL-6/anti-IL-6-receptor antibodies 71.
Another problem, specifically when using Ad and AAV vectors, is pre-existing B and T-cell responses against capsid proteins that can target infected cells or neutralize intravenously injected particles before they transduce HSCs and prevent re-administration. For example, the serum prevalence of neutralizing antibodies against AAV6 is 46% 72, and most people have serum antibodies against Ad5 and specifically against the part of Ad5 that is present in Ad5/35 73. One alternative would be to use vectors derived from Ad35, as it is one of the rarest human serotypes with a seroprevalence of <7% 74. After intravenous injection of Ad35 vectors there is only minimal transduction (only detectable by PCR) of tissues, including the liver, in human CD46 transgenic mice 75 and non-human primates 76. Ad35 vectors have been used in clinical trials as vaccine vectors. Should pre-existing immunity remain a problem transient immunosuppression has been effective when countering humoral immunity in liver gene therapy trials that used AAV8 vectors 34.
Another potential drawback for intravenous HSC gene transfer is the requirement for higher doses of gene therapy vectors when compared to ex vivo approaches due to vector sequestration by the reticulo-endothelial system and the transduction of cells other than HSCs. Also, most mobilization treatments do not specifically mobilize HSCs into the blood stream. In our studies, we have shown transduction of lineage positive cells at day 3 after HDAd5/35++ injection 65. Even though these non-HSCs could express the transgene, their half-life, compared to HSCs, is short and the potential harm of them expressing the transgene would soon be lost. As a potential solution to this problem, vectors specifically targeted to HSCs could be generated. Some work has been done in this respect 77, whereby a lentiviral vector retargeted towards CD133 efficiently transduced cells with high multi-lineage reconstituting potential. Vector sequestration by macrophages of the liver, lung and spleen also remains a major unsolved obstacle to lowering IV injected vector doses, although the impact of the requirement of higher vector titers is highly dependent on the vector platform that is being used. For example, while Ad vectors are relatively cheap and easy to produce in large amounts, this will be more challenging for lentivirus and AAV vectors.
Another group of potential problems could be associated with the mobilization process that results in a significant increase in white blood cell counts. In our mouse studies we observed up to 20-fold higher WBCs one hour after G-CSF/AMD3100 mobilization. A significant fraction of mobilized cells might resettle in the spleen and this has, in rare cases, led to splenic ruptures in patients.
In summary, in vivo HSC gene therapy has the potential to move HSC directed gene therapy out of a few very specialized centers and may enable applications for common diseases at more affordable prices. However, before a widespread application of this approach can become a reality, more rigorous efficacy and safety studies are required.
Key points.
Current protocols for hematopoietic stem gene therapy, involving the transplantation of ex vivo genetically modified HSC, are complex and not without risk for the patient.
HSCs in the bone marrow are intricately connected with the bone marrow stroma, which creates a physical barrier to transduction with intravenously injected gene transfer vectors.
Intraosseal injection of viral vectors has been shown to result in in vivo transduction of HSCs in mice. It might be more feasible and efficient in large animals and humans.
A new approach that involves the mobilization of HSCs from the bone marrow, their transduction in the periphery, and return to the bone marrow, has shown first promising results.
Footnotes
The authors declare no commercial or financial interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Zhang J, Niu C, Ye L, et al. Identification of the haematopoietic stem cell niche and control of the niche size. Nature. 2003;425(6960):836–841. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
- 2.Bonig H, Papayannopoulou T. Mobilization of hematopoietic stem/progenitor cells: general principles and molecular mechanisms. Methods Mol Biol. 2012;904:1–14. doi: 10.1007/978-1-61779-943-3_1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Trumpp A, Essers M, Wilson A. Awakening dormant haematopoietic stem cells. Nat Rev Immunol. 2010;10(3):201–209. doi: 10.1038/nri2726. [DOI] [PubMed] [Google Scholar]
- 4.Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201(8):1307–1318. doi: 10.1084/jem.20041385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Calandra G, McCarty J, McGuirk J, et al. AMD3100 plus G-CSF can successfully mobilize CD34+ cells from non-Hodgkin’s lymphoma, Hodgkin’s disease and multiple myeloma patients previously failing mobilization with chemotherapy and/or cytokine treatment: compassionate use data. Bone Marrow Transplant. 2008;41(4):331–338. doi: 10.1038/sj.bmt.1705908. [DOI] [PubMed] [Google Scholar]
- 6.Andrews RG, Bartelmez SH, Knitter GH, et al. A c-kit ligand, recombinant human stem cell factor, mediates reversible expansion of multiple CD34+ colony-forming cell types in blood and marrow of baboons. Blood. 1992;80(4):920–927. [PubMed] [Google Scholar]
- 7.Craddock CF, Nakamoto B, Andrews RG, Priestley GV, Papayannopoulou T. Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice. Blood. 1997;90(12):4779–4788. [PubMed] [Google Scholar]
- 8.Ramirez P, Rettig MP, Uy GL, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood. 2009;114(7):1340–1343. doi: 10.1182/blood-2008-10-184721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Bonig H, Watts KL, Chang KH, Kiem HP, Papayannopoulou T. Concurrent blockade of alpha4-integrin and CXCR4 in hematopoietic stem/progenitor cell mobilization. Stem Cells. 2009;27(4):836–837. doi: 10.1002/stem.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cao B, Zhang Z, Grassinger J, et al. Therapeutic targeting and rapid mobilization of endosteal HSC using a small molecule integrin antagonist. Nat Commun. 2016;7:11007. doi: 10.1038/ncomms11007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Rogers FA, Vasquez KM, Egholm M, Glazer PM. Site-directed recombination via bifunctional PNA-DNA conjugates. Proc Natl Acad Sci U S A. 2002;99(26):16695–16700. doi: 10.1073/pnas.262556899. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bahal R, Ali McNeer N, Quijano E, et al. In vivo correction of anaemia in beta-thalassemic mice by gammaPNA-mediated gene editing with nanoparticle delivery. Nat Commun. 2016;7:13304. doi: 10.1038/ncomms13304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Frecha C, Fusil F, Cosset FL, Verhoeyen E. In vivo gene delivery into hCD34+ cells in a humanized mouse model. Methods Mol Biol. 2011;737:367–390. doi: 10.1007/978-1-61779-095-9_15. [DOI] [PubMed] [Google Scholar]
- 14.Russell DW, Miller AD. Foamy virus vectors. Journal of Virology. 1996;70(1):217–222. doi: 10.1128/jvi.70.1.217-222.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Trobridge GD, Miller DG, Jacobs MA, et al. Foamy virus vector integration sites in normal human cells. Proceedings of the National Academy of Sciences. 2006;103(5):1498–1503. doi: 10.1073/pnas.0510046103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hendrie PC, Huo Y, Stolitenko RB, Russell DW. A rapid and quantitative assay for measuring neighboring gene activation by vector proviruses. Molecular Therapy. 2008;16(3):534–540. doi: 10.1038/sj.mt.6300398. [DOI] [PubMed] [Google Scholar]
- 17.Kiem HP, Allen J, Trobridge G, et al. Foamy-virus-mediated gene transfer to canine repopulating cells. Blood. 2007;109(1):65–70. doi: 10.1182/blood-2006-04-016741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Everson EM, Olzsko ME, Leap DJ, Hocum JD, Trobridge GD. A comparison of foamy and lentiviral vector genotoxicity in SCID-repopulating cells shows foamy vectors are less prone to clonal dominance. Mol Ther Methods Clin Dev. 2016;3:16048. doi: 10.1038/mtm.2016.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Henthorn PS, Somberg RL, Fimiani VM, Puck JM, Patterson DF, Felsburg PJ. IL-2R gamma gene microdeletion demonstrates that canine X-linked severe combined immunodeficiency is a homologue of the human disease. Genomics. 1994;23(1):69–74. doi: 10.1006/geno.1994.1460. [DOI] [PubMed] [Google Scholar]
- 20.Gougeon ML, Drean G, Le Deist F, et al. Human severe combined immunodeficiency disease: phenotypic and functional characteristics of peripheral B lymphocytes. J Immunol. 1990;145(9):2873–2879. [PubMed] [Google Scholar]
- 21.Burtner CR, Beard BC, Kennedy DR, et al. Intravenous injection of a foamy virus vector to correct canine SCID-X1. Blood. 2014;123(23):3578–3584. doi: 10.1182/blood-2013-11-538926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wang X, Shin SC, Chiang AF, et al. Intraosseous delivery of lentiviral vectors targeting factor VIII expression in platelets corrects murine hemophilia A. Mol Ther. 2015;23(4):617–626. doi: 10.1038/mt.2015.20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ide LM, Gangadharan B, Chiang KY, Doering CB, Spencer HT. Hematopoietic stem-cell gene therapy of hemophilia A incorporating a porcine factor VIII transgene and nonmyeloablative conditioning regimens. Blood. 2007;110(8):2855–2863. doi: 10.1182/blood-2007-04-082602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kuether EL, Schroeder JA, Fahs SA, et al. Lentivirus-mediated platelet gene therapy of murine hemophilia A with pre-existing anti-factor VIII immunity. J Thromb Haemost. 2012;10(8):1570–1580. doi: 10.1111/j.1538-7836.2012.04791.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Wang CX, Sather BD, Wang X, et al. Rapamycin relieves lentiviral vector transduction resistance in human and mouse hematopoietic stem cells. Blood. 2014;124(6):913–923. doi: 10.1182/blood-2013-12-546218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mazurier F, Doedens M, Gan OI, Dick JE. Rapid myeloerythroid repopulation after intrafemoral transplantation of NOD-SCID mice reveals a new class of human stem cells. Nat Med. 2003;9(7):959–963. doi: 10.1038/nm886. [DOI] [PubMed] [Google Scholar]
- 27.Wang J, Kimura T, Asada R, et al. SCID-repopulating cell activity of human cord blood-derived CD34- cells assured by intra-bone marrow injection. Blood. 2003;101(8):2924–2931. doi: 10.1182/blood-2002-09-2782. [DOI] [PubMed] [Google Scholar]
- 28.van Os R, Ausema A, Dontje B, van Riezen M, van Dam G, de Haan G. Engraftment of syngeneic bone marrow is not more efficient after intrafemoral transplantation than after traditional intravenous administration. Exp Hematol. 2010;38(11):1115–1123. doi: 10.1016/j.exphem.2010.07.003. [DOI] [PubMed] [Google Scholar]
- 29.Lange S, Steder A, Killian D, et al. Engraftment Efficiency after Intra-Bone Marrow Versus Intravenous Transplantation of Bone Marrow Cells in a Canine Nonmyeloablative Dla-Identical Transplantation Model. Biol Blood Marrow Transplant. 2016 doi: 10.1016/j.bbmt.2016.10.025. [DOI] [PubMed] [Google Scholar]
- 30.Feng Q, Chow PK, Frassoni F, et al. Nonhuman primate allogeneic hematopoietic stem cell transplantation by intraosseus vs intravenous injection: Engraftment, donor cell distribution, and mechanistic basis. Exp Hematol. 2008;36(11):1556–1566. doi: 10.1016/j.exphem.2008.06.010. [DOI] [PubMed] [Google Scholar]
- 31.Rocha V, Labopin M, Ruggeri A, et al. Unrelated cord blood transplantation: outcomes after single-unit intrabone injection compared with double-unit intravenous injection in patients with hematological malignancies. Transplantation. 2013;95(10):1284–1291. doi: 10.1097/TP.0b013e318288ca4d. [DOI] [PubMed] [Google Scholar]
- 32.Massollo M, Podesta M, Marini C, et al. Contact with the bone marrow microenvironment readdresses the fate of transplanted hematopoietic stem cells. Exp Hematol. 2010;38(10):968–977. doi: 10.1016/j.exphem.2010.06.003. [DOI] [PubMed] [Google Scholar]
- 33.Kay MA, Manno CS, Ragni MV, et al. Evidence for gene transfer and expression of factor IX in haemophilia B patients treated with an AAV vector. Nat Genet. 2000;24(3):257–261. doi: 10.1038/73464. [DOI] [PubMed] [Google Scholar]
- 34.Nathwani AC, Reiss UM, Tuddenham EG, et al. Long-term safety and efficacy of factor IX gene therapy in hemophilia B. N Engl J Med. 2014;371(21):1994–2004. doi: 10.1056/NEJMoa1407309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Song L, Kauss MA, Kopin E, et al. Optimizing the transduction efficiency of capsid-modified AAV6 serotype vectors in primary human hematopoietic stem cells in vitro and in a xenograft mouse model in vivo. Cytotherapy. 2013;15(8):986–998. doi: 10.1016/j.jcyt.2013.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wang J, Exline CM, DeClercq JJ, et al. Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat Biotechnol. 2015;33(12):1256–1263. doi: 10.1038/nbt.3408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Song L, Li X, Jayandharan GR, et al. High-efficiency transduction of primary human hematopoietic stem cells and erythroid lineage-restricted expression by optimized AAV6 serotype vectors in vitro and in a murine xenograft model in vivo. PLoS One. 2013;8(3):e58757. doi: 10.1371/journal.pone.0058757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Khan IF, Hirata RK, Russell DW. AAV-mediated gene targeting methods for human cells. Nat Protoc. 2011;6(4):482–501. doi: 10.1038/nprot.2011.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Khan IF, Hirata RK, Russell DW. AAV-mediated gene targeting methods for human cells. Nat Protoc. 2011;6(4):482–501. doi: 10.1038/nprot.2011.301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Maina N, Han Z, Li X, et al. Recombinant self-complementary adeno-associated virus serotype vector-mediated hematopoietic stem cell transduction and lineage-restricted, long-term transgene expression in a murine serial bone marrow transplantation model. Hum Gene Ther. 2008;19(4):376–383. doi: 10.1089/hum.2007.143. [DOI] [PubMed] [Google Scholar]
- 41.Wang J, Exline CM, DeClercq JJ, et al. Homology-driven genome editing in hematopoietic stem and progenitor cells using ZFN mRNA and AAV6 donors. Nat Biotechnol. 2015;33(12):1256–1263. doi: 10.1038/nbt.3408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.De Ravin SS, Reik A, Liu PQ, et al. Targeted gene addition in human CD34(+) hematopoietic cells for correction of X-linked chronic granulomatous disease. Nat Biotechnol. 2016;34(4):424–429. doi: 10.1038/nbt.3513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gregorevic P, Blankinship MJ, Allen JM, et al. Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nature medicine. 2004;10(8):828–834. doi: 10.1038/nm1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Gaggar A, Shayakhmetov D, Lieber A. CD46 is a cellular receptor for group B adenoviruses. Nature Medicine. 2003;9:1408–1412. doi: 10.1038/nm952. [DOI] [PubMed] [Google Scholar]
- 45.Marie JC, Astier AL, Rivailler P, Rabourdin-Combe C, Wild TF, Horvat B. Linking innate and acquired immunity: divergent role of CD46 cytoplasmic domains in T cell induced inflammation. Nat Immunol. 2002;3(7):659–666. doi: 10.1038/ni810. [DOI] [PubMed] [Google Scholar]
- 46.Li L, Krymskaya L, Wang J, et al. Genomic editing of the HIV-1 coreceptor CCR5 in adult hematopoietic stem and progenitor cells using zinc finger nucleases. Mol Ther. 2013;21(6):1259–1269. doi: 10.1038/mt.2013.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Shayakhmetov DM, Papayannopoulou T, Stamatoyannopoulos G, Lieber A. Efficient gene transfer into human CD34(+) cells by a retargeted adenovirus vector. J Virol. 2000;74(6):2567–2583. doi: 10.1128/jvi.74.6.2567-2583.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Yotnda P, Onishi H, Heslop HE, et al. Efficient infection of primitive hematopoietic stem cells by modified adenovirus. Gene Ther. 2001;8(12):930–937. doi: 10.1038/sj.gt.3301488. [DOI] [PubMed] [Google Scholar]
- 49.Lu ZZ, Ni F, Hu ZB, et al. Efficient gene transfer into hematopoietic cells by a retargeting adenoviral vector system with a chimeric fiber of adenovirus serotype 5 and 11p. Exp Hematol. 2006;34(9):1171–1182. doi: 10.1016/j.exphem.2006.05.005. [DOI] [PubMed] [Google Scholar]
- 50.Nilsson M, Karlsson S, Fan X. Functionally distinct subpopulations of cord blood CD34+ cells are transduced by adenoviral vectors with serotype 5 or 35 tropism. Mol Ther. 2004;9(3):377–388. doi: 10.1016/j.ymthe.2003.12.014. [DOI] [PubMed] [Google Scholar]
- 51.Di Paolo N, Ni S, Gaggar A, et al. Evaluation of adenovirus vectors containing seroytpe 35 fibers for tumor targeting. Cancer Gene Ther. 2006;13:1072–1081. doi: 10.1038/sj.cgt.7700981. [DOI] [PubMed] [Google Scholar]
- 52.Ni S, Bernt K, Gaggar A, Li ZY, Kiem HP, Lieber A. Evaluation of biodistribution and safety of adenovirus vectors containing group B fibers after intravenous injection into baboons. Hum Gene Ther. 2005;16(6):664–677. doi: 10.1089/hum.2005.16.664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kalyuzhniy O, Di Paolo NC, Silvestry M, et al. Adenovirus serotype 5 hexon is critical for virus infection of hepatocytes in vivo. Proc Natl Acad Sci U S A. 2008;105(14):5483–5488. doi: 10.1073/pnas.0711757105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Brunetti-Pierri N, Palmer DJ, Beaudet AL, Carey KD, Finegold M, Ng P. Acute toxicity after high-dose systemic injection of helper-dependent adenoviral vectors into non-human primates. Human Gene Therapy. 2004;15:35–46. doi: 10.1089/10430340460732445. [DOI] [PubMed] [Google Scholar]
- 55.Wang H, Liu Y, Li Z, et al. In vitro and in vivo properties of adenovirus vectors with increased affinity to CD46. J Virol. 2008;82(21):10567–10579. doi: 10.1128/JVI.01308-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Morral N, Parks RJ, Zhou H, et al. High doses of a helper-dependent adenoviral vector yield supraphysiological levels of alpha1-antitrypsin with negligible toxicity. Hum Gene Ther. 1998;9(18):2709–2716. doi: 10.1089/hum.1998.9.18-2709. [DOI] [PubMed] [Google Scholar]
- 57.Balamotis MA, Huang K, Mitani K. Efficient delivery and stable gene expression in a hematopoietic cell line using a chimeric serotype 35 fiber pseudotyped helper-dependent adenoviral vector. Virology. 2004;324(1):229–237. doi: 10.1016/j.virol.2004.03.030. [DOI] [PubMed] [Google Scholar]
- 58.Wang H, Cao H, Wohlfahrt M, Kiem HP, Lieber A. Tightly regulated gene expression in human hematopoietic stem cells after transduction with helper-dependent Ad5/35 vectors. Experimental Hematology. 2008 doi: 10.1016/j.exphem.2008.01.014. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Wang H, Shayakhmetov DM, Leege T, et al. A capsid-modified helper-dependent adenovirus vector containing the beta-globin locus control region displays a nonrandom integration pattern and allows stable, erythroid-specific gene expression. J Virol. 2005;79(17):10999–11013. doi: 10.1128/JVI.79.17.10999-11013.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Wang H, Cao H, Wohlfahrt M, Kiem HP, Lieber A. Tightly regulated gene expression in human hematopoietic stem cells after transduction with helper-dependent Ad5/35 vectors. Exp Hematol. 2008;36(7):823–831. doi: 10.1016/j.exphem.2008.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Guerrero AD, Moyes JS, Cooper LJ. The human application of gene therapy to re-program T-cell specificity using chimeric antigen receptors. Chin J Cancer. 2014;33(9):421–433. doi: 10.5732/cjc.014.10100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hausl MA, Zhang W, Muther N, et al. Hyperactive sleeping beauty transposase enables persistent phenotypic correction in mice and a canine model for hemophilia B. Mol Ther. 2010;18(11):1896–1906. doi: 10.1038/mt.2010.169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Yant SR, Ehrhardt A, Mikkelsen JG, Meuse L, Pham T, Kay MA. Transposition from a gutless adeno-transposon vector stabilizes transgene expression in vivo. Nat Biotechnol. 2002;20(10):999–1005. doi: 10.1038/nbt738. [DOI] [PubMed] [Google Scholar]
- 64.Yant SR, Wu X, Huang Y, Garrison B, Burgess SM, Kay MA. High-resolution genome-wide mapping of transposon integration in mammals. Mol Cell Biol. 2005;25(6):2085–2094. doi: 10.1128/MCB.25.6.2085-2094.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Richter M, Saydaminova K, Yumul R, et al. In vivo transduction of primitive mobilized hematopoietic stem cells after intravenous injection of integrating adenovirus vectors. Blood. 2016;128(18):2206–2217. doi: 10.1182/blood-2016-04-711580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Croyle MA, Cheng X, Wilson JM. Development of formulations that enhance physical stability of viral vectors for gene therapy. Gene Ther. 2001;8(17):1281–1290. doi: 10.1038/sj.gt.3301527. [DOI] [PubMed] [Google Scholar]
- 67.Copelan EA. Hematopoietic stem-cell transplantation. N Engl J Med. 2006;354(17):1813–1826. doi: 10.1056/NEJMra052638. [DOI] [PubMed] [Google Scholar]
- 68.Atasheva S, Shayakhmetov DM. Adenovirus sensing by the immune system. Curr Opin Virol. 2016;21:109–113. doi: 10.1016/j.coviro.2016.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Suzuki M, Bertin TK, Rogers GL, et al. Differential type I interferon-dependent transgene silencing of helper-dependent adenoviral vs. adeno-associated viral vectors in vivo. Mol Ther. 2013;21(4):796–805. doi: 10.1038/mt.2012.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Seregin SS, Appledorn DM, McBride AJ, et al. Transient pretreatment with glucocorticoid ablates innate toxicity of systemically delivered adenoviral vectors without reducing efficacy. Mol Ther. 2009;17(4):685–696. doi: 10.1038/mt.2008.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Chen F, Teachey DT, Pequignot E, et al. Measuring IL-6 and sIL-6R in serum from patients treated with tocilizumab and/or siltuximab following CAR T cell therapy. J Immunol Methods. 2016;434:1–8. doi: 10.1016/j.jim.2016.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Boutin S, Monteilhet V, Veron P, et al. Prevalence of serum IgG and neutralizing factors against adeno-associated virus (AAV) types 1, 2, 5, 6, 8, and 9 in the healthy population: implications for gene therapy using AAV vectors. Hum Gene Ther. 2010;21(6):704–712. doi: 10.1089/hum.2009.182. [DOI] [PubMed] [Google Scholar]
- 73.Bradley RR, Maxfield LF, Lynch DM, Iampietro MJ, Borducchi EN, Barouch DH. Adenovirus serotype 5-specific neutralizing antibodies target multiple hexon hypervariable regions. J Virol. 2012;86(2):1267–1272. doi: 10.1128/JVI.06165-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Barouch DH, Kik SV, Weverling GJ, et al. International seroepidemiology of adenovirus serotypes 5, 26, 35, and 48 in pediatric and adult populations. Vaccine. 2011;29(32):5203–5209. doi: 10.1016/j.vaccine.2011.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sakurai F, Kawabata K, Koizumi N, et al. Adenovirus serotype 35 vector-mediated transduction into human CD46-transgenic mice. Gene Ther. 2006;13(14):1118–1126. doi: 10.1038/sj.gt.3302749. [DOI] [PubMed] [Google Scholar]
- 76.Sakurai F, Nakamura S, Akitomo K, et al. Transduction properties of adenovirus serotype 35 vectors after intravenous administration into nonhuman primates. Mol Ther. 2008;16(4):726–733. doi: 10.1038/mt.2008.19. [DOI] [PubMed] [Google Scholar]
- 77.Brendel C, Goebel B, Daniela A, et al. CD133-targeted gene transfer into long-term repopulating hematopoietic stem cells. Mol Ther. 2015;23(1):63–70. doi: 10.1038/mt.2014.173. [DOI] [PMC free article] [PubMed] [Google Scholar]


