NZ623092B2 - Viral vectors purification system - Google Patents

Viral vectors purification system Download PDF

Info

Publication number
NZ623092B2
NZ623092B2 NZ623092A NZ62309212A NZ623092B2 NZ 623092 B2 NZ623092 B2 NZ 623092B2 NZ 623092 A NZ623092 A NZ 623092A NZ 62309212 A NZ62309212 A NZ 62309212A NZ 623092 B2 NZ623092 B2 NZ 623092B2
Authority
NZ
New Zealand
Prior art keywords
viral vector
ligand
cell surface
vector
viral
Prior art date
Application number
NZ623092A
Other versions
NZ623092A (en
Inventor
Chiara Bovolenta
Anna Stornaiuolo
Original Assignee
Molmed Spa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Molmed Spa filed Critical Molmed Spa
Priority claimed from PCT/EP2012/069713 external-priority patent/WO2013050523A1/en
Publication of NZ623092A publication Critical patent/NZ623092A/en
Publication of NZ623092B2 publication Critical patent/NZ623092B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13051Methods of production or purification of viral material
    • C12N2740/13052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/15011Lentivirus, not HIV, e.g. FIV, SIV
    • C12N2740/15051Methods of production or purification of viral material
    • C12N2740/15052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16051Methods of production or purification of viral material
    • C12N2740/16052Methods of production or purification of viral material relating to complementing cells and packaging systems for producing virus or viral particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof

Abstract

Disclosed is a method for the purification of a viral vector comprising: - introducing an exogenous gene encoding a eukaryotic cell surface receptor marker and a gene of interest in a packaging cell line, thus obtaining a producer cell line; - culturing the so obtained producer cell line, so that viral vector particles are released in the supernatant; - collecting the supernatant containing viral vector particles bearing the cell surface receptor marker on their external envelope; - incubating said supernatant with a ligand able to bind to the cell surface receptor marker thus forming a complex ligand-viral vector; - separating complex ligand-viral vector; and - obtaining purified viral vector from complex ligand-viral vector. t viral vector particles are released in the supernatant; - collecting the supernatant containing viral vector particles bearing the cell surface receptor marker on their external envelope; - incubating said supernatant with a ligand able to bind to the cell surface receptor marker thus forming a complex ligand-viral vector; - separating complex ligand-viral vector; and - obtaining purified viral vector from complex ligand-viral vector.

Description

Viral vectors purification system Field of the invention The present invention relates generally to a method for efficient purification of viral vectors (VV) ularly those belonging to the iridae family. More particularly the invention relates lly to the purification of VV by an immunological method based on the expression of an exogenous cell surface marker in the packaging cell line.
Background Viral vectors are commonly used to deliver genetic material into target cells. Nowadays VV are used in gene therapy applications to vehicle therapeutic genes into patients. In clinical applications, it is necessary to develop high quality VV in order to meet requisites imposed by regulatory agencies. Particularly, it is ary to develop safer producer cell lines, to be used in large-scale production processes in order to obtain large viral stocks. In the meantime, fficient and scalable purification ses are essential for the production of al grade viral particles to be stered in humans.
Purification of VV preparations is mandatory to prevent ty, inflammation or immune response due to vector components, cellular and medium contaminants such as for example serum (Baekland et al., 2003; Tuschong et al., 2002). Ideally a purification process needs to assure nance of viral infectivity (stability), high recovery of viral particles, removal of contaminants such as DNA, proteins and inhibitors of transduction, possibility to concentrate viral supernatant and, of , scalability of the process (Andreatis et al., 1999; Lyddiatt and O’Sullivan, 1998).
As of today, different procedures for the purification of retroviral vectors have been developed based on ent technologies, particularly: centrifugation based methods, membrane separation processes, chromatographic or other methods based on precipitation with salts and polymers such as PEG. Currently proposed cation schemes result in low recovery (approximately 30%) (Rodrigues et al., 2007). All these methods have been developed originally for protein production and, further, they have been adapted to the purification of VV. Due to the peculiarity and complexity of viral particles, it is necessary to improve purification methods in order to obtain high tivity and high hput while maintaining the biological activity of the final product, particularly in terms of ivity.
A further and more recent example is reported in Merten et al., 2011 that disclose a process for the production of lentiviral vectors in large scale and under GMP conditions, to be used in the context of a pilot gene therapy clinical trial for the treatment of Wiskott- Aldrich syndrome. The disclosed process includes both tion and downstream processes for the purification of lentiviral particles. Particularly, the purification is based on a multistep scheme combining several chromatographic and membrane based process steps including anion exchange and size exclusion chromatography. Notwithstanding the very good results in respect to the production rate and the absence of DNA and protein contaminants in the final preparations, the final yield of the purification process is in the range of previously disclosed methods (below 30%) and the ivity of the viral vectors in the final sample is reduced.
Both viral and cellular proteins are incorporated into the viral envelope during viral maturation and release from host cells and, in particular during the so called budding process (Arthur et al., 1992). It has been shown, for example, that numerous endogenous host cell proteins are incorporated into the HIV-1 envelope including human lymphocyte ns, (HLA) classes I and II, CD44, ment control proteins and others whereas others, such as CXCR4, CCR5 and CCR3, are excluded. On the basis of this observation, it was suggested that cell type specific antigens may serve as marker of the cellular origin of HIV-1 replication (Roberts et al., 1999). Furthermore, it was developed an immunomagnetic viral capture assay that was able to distinguish between lymphocytes derived and macrophages derived ated HIV viruses (Lawn et al., 2000). Particularly, Lawn et al. showed that it was le to isolate T-cell-derived HIV viruses using an antibody able to bind CD26, and to discriminate it from macrophage-derived HIV-1 viruses that, in turn, can be captured using anti CD36 dies. Both CD26 and CD36 are endogenous host cell proteins that are overexpressed during HIV-1 infection in T-cells and macrophages, tively. Both proteins are also incorporated in the viral pe thus allowing selective isolation of the virus. Lawn et al. tested a panel of antibodies able to bind host cell specific antigens before identifying the sful ones. Interestingly, several dies able to bind antigens endogenously expressed at high level by macrophages (CD32, CD64, CD88 and CD89) are instead not able to e the virus, thus showing that over-expression of a certain marker on the surface of the host cell is a necessary but not sufficient ion for capturing the virus. Lawn et al. do not show that exogenous proteins expressed by the host can be successfully orated into the virus envelope, and subsequently used for the purification of functionally active viral particles.
It has been shown that certain modified cell surface markers can be used for the purification of transduced cells. Particularly, WO/ 9506723 discloses a process of marking eukaryotic (mammalian) cells by expressing in these cells the nucleic acid encoding a cell surface receptor, that is further presented at the cell surface. This cell selection process is characterized by the use of a c acid in which the region encoding the intracellular domain of the receptor is completely or partly deleted, or modified so that the receptor presented at the surface cannot effect any signal transduction after binding to its binding partner. The cell surface receptor employed in the disclosed process is the Low Affinity Nerve Growth Factor Receptor (LNGFR), in a truncated form in which the intracellular domain has been deleted. The resulting ted cell surface receptor is called DLNGFR.
The presence of the DLNGFR n allows the in vitro immunoselection of the genetically modified cells through the use of monoclonal antibodies and ic beads.
DLNFGR is a truncated cell surface marker that is currently ed in gene therapy for the selection of transduced cells. For example, it is employed in the HSV-TK gene therapy ch, which enables safe haploidentical haematopoietic stem cell transplantation (HSCT) for the treatment of haematological malignancies. The TK therapy employs a retroviral vector which carries both the suicide gene HSV-TK and the marker gene DLNGFR (Verzeletti et al. 1998).
So far, the DLNGF receptor has not been employed for the cation of VV.
Due to the necessity of producing purified VV for clinical applications, several attempts have been made to obtain efficient cation processes that allow good recovery of VV as well as generation of sufficiently safe vectors that still have good quality in terms of ity.
The methods which are currently employed, allow to obtain low recovery and have some limits in any case, since they derive from downstream processes developed for the production of recombinant proteins and adapted to VV purification. Therefore, there is a need of efficient, fast and le cation methods for VV to be used for large scale production of vectors for gene therapy, that allow to obtain good recovery and safe viral particles which maintain high infectivity.
Summary of the invention The present invention relates generally to the field of purification of VV, particularly those belonging to the Retroviridae family. Downstream processes currently employed for the purification of VV are based on s usually applied for recombinant ns. VV are peculiar particles that are employed in basic research and gene therapy al trials and that, in the latter case, need al grade production. Purification is therefore mandatory but, due to the peculiarity of VV, there are several necessities that need to be satisfied. The purification process needs to be efficient and fast since VV are sensitive to environmental conditions, and needs to be scalable since large batches are required in clinical applications.
The present invention provides a new strategy for the purification of viral particles, that is based on the exploitation of the property of such particles to incorporate host cell proteins ed in the cellular plasma membrane into their external envelope and/or that at least provides the public with a useful choice. The purification method consists in the expression of an exogenous gene encoding a cell surface marker in the packaging cell line for the production of the VV. Such marker is exposed on the cellular membrane of the packaging cells. In the course of the production of the viral particles, during the maturation phase, the cell e marker is orated into the viral pe through the budding process.
When incorporated into the viral envelope, the maker is, in fact, a viral surface marker, but we shall continue to refer to said marker as “cell surface marker”. The viral les can be then incubated with an antibody able to recognize such marker and purified h immunological methods. All cell surface proteins that are exogenous to the packaging cells, particularly to packaging epithelial cells, can be employed as cell surface markers in the present invention. Cell surface s that can be employed are for example CD26, CD36, CD44, CD3, CD25 and the Low Nerve Growth Factor Receptor in which the intracellular domain has been deleted (DLNGFR). In a red embodiment the cell surface marker that is used for the purification process is DLNGFR.
The developed cation method is extremely ile since it is applicable to any VV that incorporates the proteins of the host cell membrane during the maturation through the budding process, such as retroviruses, lentiviruses, alpha viruses [e.g. Semliki Forest virus (SFV), Sindibis virus (SIN)], rhabdoviruses [e.g. vesicular stomatitis virus , and orthomyxoviruses [e.g. influenza A virus]. Moreover, the purification method is linked to the production method because it requires expression of the marker in the packaging cell line and, ore, allows an integrated approach in which production and downstream processes are built on the same starting material (the packaging cell line). In this context, it is le to produce stable packaging cell lines containing all elements necessary for the tion of VV as well as a further exogenous gene encoding the cell surface marker necessary for the purification. This aspect is particularly useful for both scalability and efficiency.
The method is based on the use of a ligand able to bind the cell surface marker, in order to separate VV from supernatant. Preferably the ligand is an antibody and the separation of viral particles is obtained by immunological methods. More preferably the method employs immunomagnetic selection. The method of the invention can be easily scaled-up and automated since several instruments exist for the performance of immunomagnetic selection.
Moreover the proposed method is handily, very fast and extremely efficient: the purification efficiency is higher than that obtained with the currently used s, for e chromatographic methods such as those employing DEAE and SEC columns.
In addition, it has been found that the purification method of the invention allows high recovery, since it has been shown that the titer yield of vectors purified through this method is at least 85% or even higher (120%) in most experiments in small scale. Moreover, a considerable increase of infectivity of iral vectors purified with the method of the invention has been ed (43% and 60% for transient and stable production, respectively), in small scale experiments. Such titer yield and infectivity increase are achieved with the single main step of the purification method of the invention i.e. the separation of the complex viral vector-ligand, t taking into consideration other steps that could affect the final results.
Large scale experiments have also been performed and very good results have been obtained. In fact, the recovery in terms of virus titer is 60%, with the single step of separation of the complex viral vector-ligand. Before starting the separation phase, the viral supernatant is enriched in its viral titer through preliminary filtration and centrifugation steps, that roughly eliminate contaminants and transduction inhibitors, and also through the incubation with the ligand of the receptor. The final titer yield after the te multi-step purification process (harvest viral supernatant vs final purified product) results to be more than 100%. These are very good s ering that the titer yield of the complete cation processes disclosed in the literature is around 30% (Rodrigues et al. 2007) or even below in the case of large scale preparations (Merten et al. 2011). Moreover, VV ed according to the method of the invention on a large scale result to have a three times increase of the infectivity, an even higher increase than that obtained with the same method on a small scale. This is a very important and unexpected advantage since the literature describes a se of infectivity further to the purification with conventional methods (Merten et al. 2011).
Statements of the invention In one aspect, the present invention s to a method for the purification of a viral vector comprising: i. Introducing an exogenous gene encoding a eukaryotic cell surface receptor marker and a gene of interest in a packaging cell line, thus obtaining a producer cell line ii. culturing the so obtained producer cell line, so that viral vector particles are released in the supernatant iii. collecting the supernatant containing viral vector particles bearing the cell surface receptor marker on their al envelope iv. incubating said supernatant with a ligand able to bind to the cell e receptor marker thus forming a x ligand-viral vector v. ting complex ligand-viral vector vi. obtaining purified viral vector from complex ligand-viral vector .
Certain statements that appear below are broader than what appears in the statements of the invention above. These statements are provided in the interests of ing the reader with a better understanding of the invention and its practice. The reader is directed to the anying claim set which defines the scope of the invention.
Also described herein is a method for the purification of a viral vector comprising: i. introducing an exogenous gene encoding a cell surface marker and a gene of interest (GOI) in a ing cell line ii. ing the so obtained producer cell line iii. collecting the supernatant containing viral vector particles bearing the cell surface marker on their external envelope iv. incubating said supernatant with a ligand able to bind to the cell surface marker v. ting complex ligand-viral vector vi. obtaining purified viral vector particles In another ment the supernatant containing viral vector particles g the cell surface marker on their external envelope is filtered, optionally concentrated and then incubated with a ligand able to bind to the cell surface marker.
Preferably the viral vector is a retroviral vector, a lentiviral vector, an alpha viral vector [e.g. a vector obtained from Semliki Forest virus (SFV), Sindibis virus (SIN)], a rhabdoviral vector [e.g. a vector derived from vesicular stomatitis virus (VSV)], and an orthomyxoviral vector [e.g. a vector derived from influenza A . More preferably the viral vector is a lentiviral vector or a retroviral .
In one embodiment the cell surface marker is any cell surface marker that is exogenous to the packaging cell line, which is, preferably, an epithelial packaging cell line. Preferably the cell surface marker is selected from CD26, CD36, CD44, CD3, CD25 and DLNGFR.
More preferably the cell surface marker is DLNGFR.
In one embodiment the expression of the cell surface marker is transient.
In another embodiment the expression of the cell surface marker is stable.
In one embodiment the GOI and the cell surface marker are expressed in the same transfer vector.
In another embodiment the GOI and the cell surface marker are expressed in separate vectors.
Preferably the ligand is a chemical or a biological entity selected from an agonist, an antagonist, a peptide, a peptidomimetic, an antibody, an antibody fragment, an affibody.
In a further embodiment the ligand is linked to a moiety that can be separated from the supernatant.
Preferably the ligand is an antibody.
More preferably the antibody is conjugated to magnetic beads and separation is obtained by applying a magnetic field to a on containing the complex antibody-viral vector.
Preferably the viral vectors are obtained by removing the ic field. More preferably the tion of the complex antibody-viral vector is performed on a column and the viral vectors are ed by removing the magnetic field and further eluting them from the column.
In one embodiment the viral vectors are separated from the antibody by cleaving the cell surface marker-antibody bond.
Also described herein is an exogenous cell surface marker expressed in a packaging cell line for use in the purification of viral vectors produced by said ing cell line.
Preferably the viral vector is a retroviral vector, a lentiviral vector, an alpha viral vector [e.g. a vector ed from i Forest virus (SFV), Sindibis virus (SIN)], a viral vector [e.g. a vector derived from lar stomatitis virus (VSV)], and an orthomyxoviral vector [e.g. a vector derived from influenza A virus]. More preferably the viral vector is a lentiviral vector or a retroviral vector.
The cell surface marker is exogenous to the packaging cell line, preferably exogenous to epithelial ing cells. Preferably the cell surface marker is selected from CD26, CD36, CD44, CD3, CD25 and DLNGFR.
More preferably the cell surface marker is DLNGFR.
In one ment the expression of the cell surface marker is transient.
In another ment the expression of the cell surface marker is stable.
Detailed description of the invention A detailed description of preferred features and embodiments of the invention will be described by way of non-limiting example.
The invention can be put into practice by a person of ordinary skill in the art who will employ, unless otherwise indicated, conventional techniques of chemistry, molecular biology, microbiology, recombinant DNA and immunology. All such techniques are disclosed and explained in published literature. See, for example, J. Sambrook, E. F. Fritsch, and T.
Maniatis, 1989, Molecular Cloning: A Laboratory Manual, Second Edition, Books 1-3, Cold Spring Harbor Laboratory Press; Ausubel, F.M. et al. (1995 and periodic supplements; Current Protocols in Molecular Biology, ch. 9, 13, and 16, John Wiley & Sons, New York, N.Y.); t Protocols in Immunology, ch. 12, John Wiley & Sons, New York, N.Y.); B. Roe, J.
Crabtree, and A. Kahn, 1996, DNA Isolation and Sequencing: Essential ques, John Wiley & Sons; J . M. Polak and James O'D. McGee, 1990, In Situ ization: Principles and Practice; Oxford University Press; M. J . Gait (Editor), 1984, Oligonucleotide Synthesis: A Practical Approach, IrI Press; and, D. M. J. Lilley and J. E. rg, 1992, Methods of Enzymology: DNA ure Part A: Synthesis and Physical.
Analysis of DNA Methods in Enzymology, Academic Press. All these publications are incorporated by reference.
Purification method Described herein is a new purification method for VV. ably described is a method for the purification of VV including gamma retroviruses (prototype: Moloney murine leukemia virus, Mo-MLV), lentiviruses (prototype: HIV), alpha viruses [e.g. Semliki Forest virus (SFV), Sindibis virus (SIN)], rhabdoviruses [e.g. vesicular stomatitis virus (VSV)], and orthomyxoviruses [influenza A virus]. The proposed purification method is based on one of the phases of maturation of viruses. Viral particles are ed from host cells h the budding s by which process viral capsid is wrapped with the plasma membrane derived from virus producer cells. In doing so, s incorporate in their external envelope several host cell proteins which are normally embedded in the cellular plasma membrane.
The VV produced by either a transient or stable packaging system are released from the packaging cells in an identical manner. The method of the present invention is based on the hypothesis that VV can be ically purified by using an antibody ed against an exogenous host plasma membrane n.
Described herein is a method for purification of a VV, that is based on the expression of an exogenous gene encoding a cell surface marker in a packaging cell line. The cell surface marker is a protein exogenous to the packaging cells that is expressed on the cellular membrane. Once expressed, such marker is mounted on the packaging cell membrane and, therefore, during the tion of the VV, is mounted on the external envelope of the VV ed by said ing cells. According to the method described, the supernatant containing viral particles embedding a cell surface marker on their envelope is collected and incubated with a ligand able to recognize such marker. Optionally, particularly for the purification of VV on a large scale where large volumes have to be handled and purified, the supernatant is filtered and concentrated, and then re-suspended and incubated with the ligand. All these preliminary steps allow a rough removal of contaminants and transduction inhibitors and, therefore, contribute to the enrichment in viral titer observed in the intermediate preparations and, consequently, to the final titer of the purified viral particles. r to these preliminary steps, the complex ligand-VV is separated from the medium and VV are then ed. The separation phase is the main step of the purification method as described herein. The VV can be separated from the ligand upon cleavage of the bond between the ligand and the cell surface marker.
In a preferred embodiment the ligand is an antibody.
The method of the t invention can be summarized in five main phases: 1) Expression of a cell surface marker in a packaging cell line for VV 2) tion of viral particles 3) Incubation with a ligand able to recognize the cell surface marker 4) Separation of the x ligand-receptor ) Recovery of purified viral particles Expression of the marker and production of viral particles The purification method of the present invention is based on the expression of an ous cell surface marker in the packaging cell line for the VV. In a preferred embodiment the cell surface marker is ous to epithelial ing cells. Preferably the cell surface marker is selected from CD26, CD36, CD44, CD3, CD25 and the truncated form of Low Nerve Growth Factor Receptor missing the ellular domain R). In a preferred embodiment the cell surface marker is DLNGFR. The expression of the cell surface marker can be obtained in several ways. In one embodiment, the cell surface marker can be transiently expressed in the packaging cell line. In one embodiment the cell surface marker and the eutic gene are both expressed in the same transfer . In another embodiment the cell surface marker and the therapeutic gene are expressed in separate s.
In another embodiment the expression of the cell surface marker is stable. Also described is a packaging cell line containing all structural elements necessary for the production of VV such as viral gag/pol, rev, optionally tat and the envelope protein of interest, together with the cell surface marker. In one embodiment, all these genes are stably integrated into the stable packaging cell line. In another embodiment the elements necessary for the production of VV are transiently expressed. The packaging cell line can be used to produce VV further to the introduction of the transfer vector containing the GOI. This packaging cell line ents an integrated technical solution that contains all elements necessary for the production of the VV and allows a rapid, safe and efficient cation method.
Further to the introduction of the transfer vectors, the production is obtained by culturing the packaging cell line containing the stably integrated or transiently expressed cell surface marker. The viral particles incorporate the cell surface marker into their envelope during the budding process and they are ed in the supernatant. Purified viral particles will be obtained by exploiting the presence of the exogenous cell surface marker as described above.
In another embodiment there is provided a producer cell line containing all structural elements necessary for the production of VV such as viral gag/pol, rev, optionally tat, the envelope protein of st and the GOI, together with the cell surface marker. Following to the culturing of the producer cells, viral particles containing the cell surface marker are released in the supernatant and they are purified exploiting the ce of the exogenous cell e marker as described above.
Incubation with ligand and separation of the complex ligand-cell e marker Viral particles containing a cell surface marker incorporated into their envelope can be isolated. According to the purification method of the present invention, the supernatant containing VV is incubated with the ligand able to ize the cell surface marker. In another embodiment the supernatant containing the VV is first filtered and concentrated and then is ted with the ligand. The ligand is linked to another structure that allows separation from the supernatant and, consequently, isolation of the ligand-VV complex. The ligands that can be used in the present invention are chemical or ical entities including but not limited to agonists, antagonists, es, peptidomimetics, antibodies, antibody fragments, affibodies.
Preferably the ligand is an antibody and the method of the present invention comprises immunoselection for the separation of the complex antibody-VV. In this case, VV containing the cell surface marker on their envelope may be selected on the basis of their reactivity with the ell surface marker antibodies.
More preferably, the method of the present invention comprises immunomagnetic selection. Immunomagnetic ion refers to the coupling of dies to paramagnetic microspheres (beads) enabling a separation of the antigenic structures by means of a magnet. For example, supernatant containing VV incorporating the cell e marker into their envelope may be incubated with a primary IgG anti-cell surface receptor antibody. The iral supernatant may be then incubated with immunomagnetic beads coated with anti- IgG secondary Ab, and applied to a magnet in order to te the retroviral vectors carrying the marker. After separation from retroviral supernatant, the isolated vectors may be recovered by ng the ic field. Alternatively, the anti-cell surface receptor antibodies can be directly conjugated to magnetic beads. In this case, immediately after the single incubation phase the magnetic field is d to the solution.
Paramagnetic microsphere to be employed in the present invention are known in the art, they are polymer particle having small size ranging from 50 nm such as the commercially available MACS® microbeads from Miltenyi Biotec, to bigger particles of 0.5-500 mm such as the commercially available Dynabeads®, from Invitrogen. Paramagnetic microsphere can be directly or indirectly conjugated to the specific antibody of interest able to bind the cell surface marker incorporated into the VV pe. Method of conjugation of antibody to paramagnetic beads are known in the art and include, for example, cross linking, formation of covalent bonds on functional groups, biotin-avidin system and others. tion of the VV from viral supernatant will be obtained by applying a magnetic field to a solution ning the complex consisting of the dy conjugated paramagnetic beads and linked to the cell surface marker.
In a preferred embodiment immunomagnetic selection is performed on a column.
Particularly, the supernatant containing VV may be directly ted with an antibody able to recognize the cell surface marker, such antibody being conjugated to paramagnetic beads.
In r embodiment the supernatant ning the VV is first filtered and optionally concentrated and then is incubated as previously described. Following incubation, the supernatant or the filtered and optionally concentrated solution is applied on a column placed in a ic separator for the removal of impurities and separation of the viral particles that remain in the column thanks to the magnetic field.
Recovery of purified particles According to the method of the invention, the last phase of the process is the recovery of purified viral particles. Such recovery is obtained removing the viral particles from the magnetic field. If the purification is performed on a column the recovery is obtained by removing the ic field. Viral vectors can then be separated from the ligand by cleaving the bond between the ligand and the cell surface marker. s for cleaving said bond are known in the art and include the use of displacement ligands or of appropriate solutions containing enzymes. Depending on the nature of the ligand and of the receptor and their bond the appropriate method is employed.
Efficiency, scale-up and tion The method of the invention is ely efficient and is simple and fast. Currently proposed purification methods allow a recovery around 30%. Remarkably the method of the invention allows to obtain titer yield of at least 85% or even higher (120%) in most experiments in small scale. The titer yield, in this case, has been ated referring to the main step of the method of the invention: the separation of the x viral vector-ligand. The abovementioned titer yield results from the ratio between the titer of the unpurified particles incubated with the ligand of the ous receptor and the titer of the purified particles.
The fied particles in ments on a small scale are obtained from the VV supernatant through preliminary tion step, and are then incubated with the ligand of the exogenous receptor. The results show that the recovery in terms of viral titer with the method of the invention is very high.
Viral particles are extremely labile and sensitive to environmental conditions. In particular, the presence of the cell surface maker on the envelope of VV could, in principle, affect the composition of such envelope as well as its ure and the availability of viral pe proteins, affecting in turn the tropism of the vector and, consequently, causing problems to viral titer and ivity. With the method of the invention, on the contrary, the titer is unaffected or even increased and, remarkably, the infectivity of lentiviruses purified with such method is increased (43% and 60% for transient and stable production, respectively) in small scale. These results are surprising because of the presence of structural elements that could, ly, negatively affect the tropism and the infectivity of the vectors.
A further advantage of the invention is that the method can be simply scaled-up and automated. Considering the case in which VV are purified by using immunomagnetic selection, it is possible to employ machines (e.g. CliniMacs® Plus Instrument, from Miltenyi Biotec) able to perform automated selection. Such machines must be able to perform liquid exchange on a solid support such as a column and immunomagnetic selection through the generation of a magnetic field. Automation helps the production of large VV stocks since it allows the purification of large quantity of viral supernatant. Purification of VV on a large scale with the method of the invention allows to obtain at least 60% titer yield in the main ic tion phase (titer of the unpurified particles incubated with ligand of the receptor vs titer of the purified les). The unpurified particles, in experiments on a large scale, are obtained from the VV supernatant through preliminary purification steps: filtrations, optionally concentration, and are then incubated with the ligand of the exogenous or. Each of these steps causes a rough removal of contaminants and transduction inhibitors, with a progressive enrichment in viral titer of the sample that undergoes the separation. The titer yield of the entire s on a large scale (titer of harvest viral supernatant vs titer of purified particles) results to be more than 100%. These are a very important results considering that the titer yield of an entire purification process of VV is reported to be around 30% (Rodrigues et al. 2007) or even below in the case of large scale ations (Merten et al. 2011). The purification on a large scale with the method of the present invention allows an increase of the infectivity of VV that result to be about 3 times more infective than the unpurified particles after the separation. In addition, in order to have an idea of the quality of the preparation obtained with the purification method of the t invention, it is le to count the number of the lentiviral infectious particles (Transfecting Units derivable from the viral titer) vs total physical particle (obtainable from the conventional equation that 1ng of p24Gag corresponds to 107 physical les, as ed in Salmon and Trono, 2006). It is very interesting to note that, with the method of the invention, as shown for example in Experiment 2 of Table 4, starting from a supernatant containing 1 infectious particle out of 5,318 total physical particles (a very poor ng material), it was possible to obtain a purified preparation containing 1 infectious particle out of 250 total physical particles, with a very good enrichment in terms of quality and functionality of the LV preparation.
In this specification where nce has been made to patent specifications, other external documents, or other s of information, this is generally for the purpose of providing a context for discussing the es of the invention. Unless specifically stated otherwise, reference to such external documents is not to be construed as an admission that such documents, or such sources of information, in any jurisdiction, are prior art, or form part of the common general knowledge in the art.
The term “comprising” as used in this specification and claims means “consisting at least in part of”. When interpreting ents in this specification, and claims which include the term “comprising”, it is to be understood that other features that are additional to the features prefaced by this term in each statement or claim may also be present. d terms such as “comprise” and “comprised” are to be interpreted in similar manner. pFurther preferred features and embodiments of the present invention will now be described by way of non-limiting example and with reference to the accompanying drawings in which: Description of the s Figure 1. Schematic representation of the strategy of the anti-DLNGFR-Ab-based purification process. “Constructs” indicates the plasmids or vectors required to produce either transiently or stably the VV to be purified (step 1). The cassette of the selection marker DLNGFR can be incorporated into the transfer vector construct or in a different plasmid expressed either transiently or stably from the ing cells. The VV are purified by the anti-DLNGFR Ab coupled to magnetic beads which are then retrained in a magnetic column (step 2) and y eluted (step 3). The purified VV can be either used with the attached beads (step 3.1) or after l of the ed beads (step 3.2).
Figure 2. Schematic representation of the experimental procedure of the invention. The procedure is divided in three simple steps: 1) the magnetic labelling of the VV consisting in the incubation of supernatants with the anti-DLNGFR Ab coniugated microbead suspension for 30 minutes at room temperature; 2) the magnetic separation of the VV consisting in the application of the sample into the magnetic column placed into the ic separator; all sample components (i.e contaminants, proteins and excess Abs) go in the flow-through and further removed by several ; 3) the elution of the VV consisting in the removal of the column from the magnetic separator and collection of the purified VV. The purified VV can be either used with the attached beads (step 3.1) or after removal of the attached beads (step 3.2).
Figure 3. Graph summarizing the experimental data in small scale. The yield of the VV titer has been calculated as percentage of the titer of the purified VV vs that of the magnetically labeled VV before loading the sample into the column. The values are the means of 5 experiments for the iral s, produced stably by the RD2-MolPack-Chim3.14 packaging clone, which carry the RD114-TR envelope (Stable LV, RD114-TR); 6 ments for the lentiviral s, produced by transient transfection of HEK-293T, which carry the VSV-G envelope (Trans. Transf. LV, VSV-G); 5 experiments for the retroviral vectors, produced from the AM12-SFCMM-TK clone 48, which carry an Ampho pe (Stable RV, Ampho).
Examples e I: Production of VV Stable production of MLV retroviral vectors (RV) Murine NIH-3T3-derived, e4070-pseudotyped AM12-SFCMM-TK clone 48 packaging cells were grown in DMEM (Dulbecco’s Modified Eagle Medium) ittaker™, Cambrex Bio Science Walkersville, Inc. Walkersville, MD) or X-VIVO 15 supplemented with 10% FBS (BioWhittaker™) and 2 mM glutamine at 37°C in 5% CO2 atmosphere. The AM12-SFCMM-TK clone 48 was obtained after transduction of the construct SFCMM-3 Mut2, which encodes a modified form of the HSV-TK gene characterized by a single silent mutation at nucleotide 330 of the ORF (WO 23912). Transduced cells were immune selected by using the anti-DLNGFR mAb of the Am12-SFCMM-3 Mut 2 cells and then cloned by limiting dilution (0.3 cell/well). AM12-SFCMM-TK clone 48 contains two copies of SFCMM-3 Mut2 vector. The GMP-grade retroviral vector supernatant lots were produced either in roller bottles or in a packed-bed 32-liter bioreactor in X-VIVO 15 medium in the presence of 1% glutamine, 10% PBS and isoleucine/tryptophan/Na e.
Stable production of lentiviral vectors (LV) Human HEK-293T and its derivative RD2-MolPack-Chim3 packaging cells were propagated in co’s Modified Eagle Medium (DMEM) supplemented with 10% FCS and PSG. RD2- MolPack-Chim3.14 and Chim3.25 clones stably produce second generation LV for anti-HIV gene therapy approach. The clones were obtained by sequential integration of the packaging constructs and the transfer vector using integrating vectors. Briefly, HEK-293T cells were transiently transfected with a d encoding the adeno-associated virus (AAV) Rep-78 protein and then infected with an hybrid baculovirus-AAV vector, in which the baculoviral backbone contains an integration cassette expressing the HIV-1 structural gag, pol, the tory rev and the hygro-resistance genes flanked by the AAV inverted terminal repeats (ITR) sequences national Patent Application N° ). This , which allows the Rep78-mediated ation of the ITR-flanked cassette into HEK-293T genome, generated the first intermediate clone named PK-7. From PK-7 clone, the RD2-MolPack- 14 and Chim3.25 ing clones (International Patent Application N° WO 2012/028681) were obtained through the sequential integration of the SIN-LV sing the HIV-1 regulatory tat and the chimeric RD114-TR envelope gene and the Tat-dependent LV vector expressing the IV Vif dominant negative transgene Chim3. Clones were obtained by seeding the cells at limiting dilution in 96-well plate (0.1 to 0.3 cell/well). For each cell type cloning experiment, at least 5 to 10 individual clones or more were selected by visual inspection under l microscope and lly expanded. LV derived from RD2- MolPack-Chim3 were obtained by small scale culture in either T25 or T75 flasks and by large scale in T162 flasks Transient production of LV Pseudotyped LV produced from HEK-293T cells were obtained by transient co-transfection of the following plasmids: the packaging constructs CMV-GPRT, the VSV-G construct, and the 2nd-gen. PDN-Chim3 transfer vector (International Patent Application N° ).
The ratio of packaging:envelope:transfer s corresponded to 6.5:3.5:10 mg DNA.
Transient transfections were performed with either the standard Ca2+-PO4 method or the FugeneTM6 system following the manufacturer’s instruction (Roche Diagnostics Corporation, Indianapolis, IN) obtaining similar results. Supernatants were harvested 48 hours after transfection and ed through a 0.45-μm .
Example II: Purification of VV by the anti-LNGFR Abs on a small scale Small scale purification of VV was d out as follows. Supernatants containing VV were diluted with 1:5 (vol/vol) with PBS ning 0.5% BSA and then filtered with 0.45 mm filters. From one to five ml of diluted supernatants were incubated with anti-LNGFR Ab coniugated microbeads suspension (CD271 Microbeads yi Biotec, GmbH, Germany cat. #130330) at the 1:40 ratio (vol/vol). The samples were then incubated at room temperature (RT) for 30 minutes on a rotating wheel. The magnetically labelled samples were loaded on the column placed into the magnetic separator, (Miltenyi, MS Columns cat. #130201). After the flow-through was collected for analysis and three washes were performed with 0.5 ml of washing buffer (PBS ning 2% FCS and 0.5% BSA), the column was removed from the magnetic separator and the purified VV were collected.
Example III: titer calculation VV titer was calculated on SupT1 cells by transducing them by one cycle of ulation at 1,240 × g for 1 hour in the presence of polybrene 8 μg/ml (Sigma-Aldrich, St Louis, MO). uction efficiency was monitored by flow cytometry analysis (FACS Calibur BD Bioscience, San Jose, CA) of DLNFGR expression, as described in Porcellini et al., 2009 & 2010, using the FlowJo software (Tree Star, Inc., Ashland, OR). Only transduction values ranging from 5 to 20% positive cells were used to calculate the titer according to the formula: TU = [number of cells × (% positive 100)]/vol sup (in ml).
Example IV: analysis of potency of ed versus fied VV in small scale preparation Several experiments were performed, as summarized on Table 1, and Table 2, using three types of VV produced by different modalities and pseudotyped with distinct envelopes. The 2nd generation LV expressing the Chim3 transgene were produced from either the stable packaging cell line RD2-MolPack-Chim3.14 or by transient transfection of HEK-293T cells as reported in Example I. In the first ion, the LV were pseudotyped with the chimeric RD114-TR envelope, made of the ellular and trans-membrane domains of the feline endogenous retrovirus RD114 envelope and the cytoplasmic tail (TR) of the A-MLVenv 4070A (Sandrin et al., 2002), whereas in the second condition with the vesicular stomatitis virus glycoprotein G ) envelope. The gRV were produced from the AM12-SFCMM-TK clone 48 and carried the MLV e4070 envelope.
Each experiment was carried out at the ing conditions: 1) diluted supernatant volume (one ml of supernatant diluted 1:5 with PBS/2% FCS/0.5% BSA); 2) supernatant:microbead suspension (vol:vol) ratio 1:40; 3) anti-LNGFR Abs directly coupled to magnetic beads (CD271 microbeads). The output of the analysis corresponds to both the percentage of titer yield (Figure 3A) and the percentage of the increment of infectivity (Figure 3B) of purified VV ve to that of unpurified ically labelled VV. Titer calculation was med on SupT1 cells, as detailed in Example III. Remarkably, the yield for LV produced stably was superior to 100% (121% on average) and that of LV produced transiently was 90%. This means that the purification of LV, regardless the type of envelope they mount, is highly effective in removing serum proteins or other contaminants that might decrease titer values.
The purification yield of gRV is slightly inferior to that of LV (85%). Moreover, a considerable increase of infectivity of lentiviral vectors purified with the method of the invention has been obtained (43% and 60% for transient and stable production, respectively). e V: Purification of VV by the NGFR Abs on a large scale Large scale purification of VV was carried as follows. Filtered supernatants (0.45mm) containing LV (800 ml) were concentrated 8-fold by centrifugation at low speed (3,400×g) for 16 hours at +4°C in refrigerated bench top centrifuge. VV pellets were resuspended in 100 ml buffer PBS/EDTA 0.5% human serum albumin (HSA) and then incubated with anti-LNGFR Ab coniugated eads suspension (CD271 Microbeads, Miltenyi Biotec, cat. #130 330) at the 1:40 ratio (vol/vol) in a 150-ml transfer bag (Miltenyi Biotec cat. #183-01). The s were then incubated at RT for 30 minutes on an orbital shaker. The magnetically ed samples were loaded on the CliniMacs® Plus Instrument and the automated separation programme Enrichment 3.2 was started. The purified LV was recovered in 40 ml and an aliquot was evaluated for cation performance by potency ation.
Example VI: analysis of potency of purified versus unpurified VV in large scale preparation Two experiments were performed, using the 2nd generation LV expressing the Chim3 transgene produced from the stable packaging cell line RD2-MolPack-Chim3.25. s are summarized on Table 3 and 4. Each experiment was carried out as described in Example V.
The output of the analysis corresponds to both the percentage of titer yield and the percentage of the increment of infectivity of purified LV relative to that of unpurified viral particles bound to NGFR Ab conjugated to the magnetic beads (Table 3) or relative to that of supernatant (Table 4). Titer calculation was performed on SupT1 cells as sed in example III. The titer yield of large scale purification (Table 3, EL/Input) was around 60% in the single step of separation of the complex viral -ligand. Before starting the separation phase, the viral supernatant is enriched in its viral titer through preliminary filtration and centrifugation steps, that roughly eliminate contaminants and transduction inhibitors, and also through the incubation with the ligand of the receptor. The final titer yield after the complete multi-step cation process (harvest viral supernatant vs final ed product) (Table 4, EL/Sup), is more than 100%: 118% for experiment 1 and 231% for experiment 2. Most importantly, the infectivity of purified particles is dramatically increased of three times in respect to the unpurified, with an even higher ment as compared to the small scale experiments indicating that large scale and automation further increase the yield of the process in terms of functionality of VV.
Table 1. Summary of experiments in small scale DLNGFR Type of Production Vector Env TU Yield (%)a Infect.(%)b Exp copy Stable LV LV 20 RD114-TR 121.4±24SEM 59.8±29SEM 5 Trans. Transf. LVc LV nd VSV-G 90.6±9.3SEM 43.0±12SEM 5 Stable RV RV 2 e4070 85.0±6.4SEM nd 6 iations: LV, lentiviral vector; RV, retroviral vector; RD114-TR, chimeric envelope from the feline endogenous retrovirus and the TR domain of MLV env; VSV-G, lar stomatitis virus envelope glycoprotein G aYield has been ated as the % of total TU recovered respect to the total TU input loaded into the magnetic column. bInfectivity has been calculated as the % of increment of infectivity of purified versus input VV. cTransient ection has been carried out on HEK293T cells as described in the text.
Table 2. y of all ments in small scale Stable LV (RD114-TR) Number of exp. EX1 EX2 EX3 EX4 EX5 Titera p24Gagb Infect.c Titera p24Gagb Infect.c Titera p24Gagb Infect.c Titera b Infect.c Titera p24Gagb Infect.c Input 3 1,3 3.0×103 4.5×103 1.2 3.8×103 5.2×103 1,5 3.4×103 5.2×103 1,5 3.4×103 7.9×103 1,5 5.2×103 Flow h 0 0,1 nd 1.2×103 nd nd 0 0,3 nd 1.9×103 0,28 6.8×103 3.0×102 0,3 1.0×103 Eluted 8.2×103 1,1 7.5×103 3 0,8 4.6×103 6.3×103 1,2 5.2×103 7.3×103 1,1 6.6×103 5.1×103 1,2 3 YIELD: %recd. & %var.FT/Ine ndd 7.6d nde 26 nd nd nd 20 nd 36 18 50 3.7 20 -80 YIELD: %rec.f & %var.EL/Ing 200f 84f 150g 82 66 21 121 80 53 140 73 94 64 80 -19 Transient Transfection LV (VSV-G) Number of exp. EX1 EX2 EX3 EX4 EX5 Titera p24Gagb .c Titera p24Gagb Infect.c Titera p24Gagb Infect.c Titera p24Gagb .c Titera p24Gagb Infect.c Input 1.4×107 165 8.4×104 1.3×107 160 8.1×104 1.4×107 187 7.4×104 1.7×107 196 8.6×104 1.7×107 196 8.6×104 Flow through 2.1×106 44 4.9×104 2.0×106 22 9.0×104 5.0×106 81 6.1×104 7.2×106 91 7.9×104 5.6×106 81 6.9×104 Eluted 7 101 1.6×105 1.0×107 100 1.0×105 7 110 1.0×105 1.3×107 105 1.2×105 1.2×107 108 1.1×105 YIELD: %recd. & %var.FT/Ine 15 26 -41 15 14 10 36 43 -17 42 46 -8 33 41 -20 YIELD: %rec.f & %var.EL/Ing 121 61 90 76 62 23 78 59 35 76 53 39 70 55 28 Stable RV (Ampho) Number of exp. EX1 EX2 EX3 EX4 EX5 EX6 Titer p19Gag Infect. Titer p19Gag Infect. Titer p19Gag Infect. Titer p19Gag Infect. Titer p19Gag Infect. Titer p19Gag Infect.
Input 6.0×104 8.0×104 9.3×104 9.3×104 7.5×104 7.5×104 Flow through 7.6×103 nd 5.4×103 nd 1.5×104 nd 3.0×104 nd 1.5×104 nd 1.4×104 nd Eluted 6.9×104 6.8×104 5.8×104 8.1×104 6.1×104 6.0×104 YIELD: %recd. & %var.FT/Ine 12 6.7 16 32 20 19 YIELD: %rec.f & %var.EL/Ing 115 85 62 87 81 80 aTiter of the total amount of loaded and eluted VV bTotal amount of p24Gag expressed in ng cInfectivity expressed as TU/ng p24Gag dPercentage of total titer and p24Gag in the flow thorugh material respect to the total titer and p24Gag of the input VV ePercentage of variation of the infectivity in the flow through material respect to the ivity of the input VV fPercentage of total titer and p24Gag in the eluted material respect to the total titer and p24Gag of the input VV ntage of variation of the infectivity in the eluted material respect to the infectivity of the input VV nd: not determined Table 3. Summary of large scale ments Stable LV (RD114-TR) Number of exp. EX1 EX2 Titera p24Gagb Infect.c Inf/Totd Titera p24Gagb Infect.c Inf/Totd Input (102.5 ml) 6.1×107 6,700 7.0×103 1:1,098 8.6×107 6,060 1.4×104 1:704 Eluted (40 ml) 3.9×107 1,480 4 1:358 5.2×107 1,300 4.0×104 1:250 YIELD: %rece & %var.f of EL/IN 64e 22e 371f 60e 21e 285f aTiter of the total amount of loaded and eluted LV bTotal amount of p24Gag expressed in ng cInfectivity expressed as TU/ng p24Gag dRatio between total infectious vs physical particles ePercentage of total titer and p24Gag in the eluted material respect to the total titer and p24Gag of the input LV, rec recovery fPercentage of variation of the infectivity in the eluted material respect to the infectivity of the input LV, var variation Table 4. Large scale full process yield Stable LV (RD114-TR) Number of exp. EX1 EX2 Titera p24Gagb Infect.c Inf/Totd Titera p24Gagb Infect.c Inf/Totd Sup. (800 ml) 7 11,600 2.8×103 0 2.2×107 11,700 1.8×103 1:5,318 Eluted (40 ml) 3.9×107 1,480 2.6×104 1:358 5.2×107 1,300 4.0×104 1:250 YIELD: %rece & %var.f of EL/SUP 118e 13e 928f 231e 11e 2222f aTiter of the total amount of starting and eluted LV bTotal amount of p24Gag expressed in ng tivity expressed as TU/ng p24Gag dRatio between total infectious vs physical particles ePercentage of total titer and p24Gag in the eluted material respect to the total titer and p24Gag of the input LV, rec recovery fPercentage of variation of the infectivity in the eluted material respect to the infectivity of the input LV, var variation References 1. Baekeland, V., et al. (2003) Optimized lentiviral vector tion and purification procedure prevents immune response after transduction of mouse brain. Gene Ther 10: 940. 2. Tuschong, L., et. al. (2002). Immune response to fetal calf serum by two adenosine deaminase-deficient patients after T cell gene therapy. Hum. Gene Ther. 13: 1605-1610. 3. Andreadis, S.T., et al. (1999). Large scale processing of recombinant retrovirus for gene therapy. hnolo. Prog. 15: 1-11. 4. tt,A., and O'Sullivan, D.A. (1998). Biochemical recovery and purification of gene therapy vectors. Curr. Opin Biotechnol. 9: 177-185. . ues, et al. (2007). Purification of retroviral vectors for clinical application: biological implications and technological challenges. J. of Biotech. 127: 520-541. 6. Arthur, L.O, et al. (1992). Cellular proteins bound to immunodeficiency viruses: implication for pathogenesis and es. Science 258: 1935-1938. 7. Roberts, B. D., et al. (1999). Host n incorporation is conserved among e HIV-1 subtypes. AIDS 13: 425-427. 8. Lawn, S. D., et al. (2000). Cellular compartments of human immunodeficiency virus type 1 replication in vivo: determination by presence of virion-associated host proteins and impact of opportunistic infection. J Virol 74 (1): 139-145 9. Verzeletti, S., et al. (1998). Herpes simplex virus thymidine kinase gene transfer for controlled graft-versus-host disease and graft-versus-leukemia: clinical -up and improved new vectors. Hum. Gene Ther, 9(15):2243-51 . Sandrin, V., et al. (2002). Lentiviral vectors pseudotyped with a modified RD114 envelope glycoprotein show increased stability in sera and augmented transduction of primary lymphocytes and CD34+ cells derived from human and nonhuman primates. Blood 100: 823-832. 11. Porcellini, S., et al. (2009). The F12-Vif derivative Chim3 inhibits HIV-1 replication in CD4+ T cytes and CD34+-derived macrophages by blocking HIV-1 DNA ation.
Blood 113: 3443-3452. 12. Porcellini, S., et al. (2010). Chim3 confers al advantage to CD4+ T cells upon HIV-1 infection by preventing HIV-1 DNA integration and HIVinduced G2 cell-cycle delay.
Blood 115: 4021-4029. 13. Bastiani Lallos, L., Laal, S., Hoxie, J.A., Zolla-Pazner, S., and Bandres, J.C. (1999) Exclusion of HIV coreceptor CXCR4, CCR5, and CCR3 from the HIV envelope. AIDS Research and Human iruses 15: 895-897. 14. s et al., (2011). Large-Scale manufacture and characterization of a lentiviral vector produced for clinical ex vivo gene therapy application. Hum. Gene Ther 22:343–356.
Patrick Salmon and Didier Trono, (2006) Current Protocols in Neuroscience Supplement 37: 4.21.1-4.21.24, John Wiley & Sons, Inc.

Claims (3)

Claims
1. A method for the purification of a viral vector comprising: i. ucing an exogenous gene encoding a eukaryotic cell surface receptor marker and a gene of st in a packaging cell line, thus 5 obtaining a producer cell line ii. culturing the so obtained producer cell line, so that viral vector particles are released in the supernatant iii. collecting the atant containing viral vector particles bearing the cell surface receptor marker on their external envelope 10 iv. incubating said supernatant with a ligand able to bind to the cell surface receptor marker thus forming a x ligand-viral vector v. separating complex ligand-viral vector vi. obtaining purified viral vector from complex ligand-viral vector .
2. A method according to claim 1, wherein the viral vector is selected from a retroviral 15 , a lentiviral vector, an alpha viral vector, a rhabdoviral vector, and a orthomyxoviral vector. 3. A method according to claim 1 or 2 wherein the cell surface receptor marker is selected from CD26, CD36, CD44, CD3, CD25 or the truncated form of Low Nerve Growth Factor Receptor (DLNGFR). 20 4. A method according to any one of claims 1 to 3 wherein the cell e receptor marker is the truncated form of Low Nerve Growth Factor Receptor (DLNGFR). 5. A method according to any one of claims 1 to 4, n the expression of the cell surface receptor marker is transient. 6. A method according to any one of claims 1 to 4, wherein the expression of the cell 25 surface receptor marker is stable. 7. A method according to any one of claims 1 to 6 wherein the gene of interest and the exogenous gene are sed in the same transfer vector. 8. A method according to any one of claims 1 to 6 wherein the gene of interest and the exogenous gene are sed in separate vectors. 9. A method according to any preceding claim wherein the ligand is a chemical or a biological entity selected from the group consisting of agonists, antagonists, es, peptidomimetics, antibodies, antibody fragments, and affibodies. 10. A method according to any preceding claim wherein the ligand is linked to a moiety 5 that can be separated from the supernatant. 11. A method according to claim 9 or 10 wherein the ligand is an antibody conjugated to magnetic beads and separation is obtained by applying a magnetic field to a solution containing the complex antibody-viral vector. 12. A method according to claim 11 wherein purified VV are obtained by removing the 10 magnetic field. 13. A method according to any preceding claim n the separation of the complex ligand-viral vector is performed on a column. 14. A method as defined in any one of claims 1 to 13 ntially as herein described with nce to any example thereof. 1 2 Magnetic column Constructs Viral Vector (VV) Envelope Transfer vector+ ∆LNGFR ∆LNGFR ∆LNGFR
3.1 Purified VV with 3.2 Purified VV with Packaging cell bead attached bead removed
NZ623092A 2011-10-05 2012-10-05 Viral vectors purification system NZ623092B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP11183937 2011-10-05
EP11183937.9 2011-10-05
PCT/EP2012/069713 WO2013050523A1 (en) 2011-10-05 2012-10-05 Viral vectors purification system

Publications (2)

Publication Number Publication Date
NZ623092A NZ623092A (en) 2016-07-29
NZ623092B2 true NZ623092B2 (en) 2016-11-01

Family

ID=

Similar Documents

Publication Publication Date Title
US20220243223A1 (en) Methods, kits, agents and apparatuses for transduction
CN107002084B (en) Co-stimulatory chimeric antigen receptor T cells targeting IL13R alpha 2
AU2016355323B2 (en) Buffers for stabilization of lentiviral preparations
CN116063574A (en) HER 2-targeting chimeric antigen receptor
US11957715B2 (en) Method for NK cell transduction
EP3186382B1 (en) Method and means for purifying retroviral vectors
CN115074331A (en) Chimeric antigen receptor targeting PSCA
US20140248695A1 (en) Viral Vectors Purification System
CA3110926A1 (en) Improved lentiviral vector
CN113045675A (en) Antibody for resisting CD22 protein molecule and application thereof
NZ623092B2 (en) Viral vectors purification system
TW202321443A (en) Cell lines for producing a retroviral vector encoding a car
Schatz Development of target antigen-displaying virus-like particles (VLPs) for the generation of antibodies using hybridoma technology