专利摘要:
The invention relates to a method for purifying enveloped viruses. The method of the invention is useful for large-scale recovery of enveloped viruses under conditions of good manufacturing practice and for obtaining clinical grade virus.
公开号:FR3014901A1
申请号:FR1362835
申请日:2013-12-17
公开日:2015-06-19
发明作者:Driss Boudeffa;Otto-Wilhelm Merten;David Fenard
申请人:Genethon;
IPC主号:
专利说明:

[0001] The invention relates to a method for purifying enveloped viruses. The method of the invention is useful for large-scale recovery of enveloped viruses under conditions of good manufacturing practice and for obtaining clinical grade virus. BACKGROUND TECHNOLOGY Lentiviral vectors derived from the human immunodeficiency virus (HIV-1 in particular) are among the most widely used vectors for gene therapy. These vectors are usually pseudotyped with glycoproteins from other viruses: gibbon leukemogenous virus (GALV), vesicular stomatitis virus (VSV-g), measles virus (or MV for measles virus in English). Methods for purifying clinical lots of lentiviral vectors pseudotyped with the VSV-G protein have been described (Schweizer and Merten, 2010). However, the pseudotyped viral vectors with other envelope proteins, and more particularly with glycoproteins derived from GaLV or MV, are relatively little used because no satisfactory purification protocol is available at present. The major limiting obstacle for the purification of this type of pseudotyped vector is related to the instability and fragility of certain membrane glycoproteins. However, these vectors are particularly interesting with regard to their tropism less wide than that of vectors pseudotyped with the VSV-G protein. For example, vectors pseudotyped with a glycoprotein derived from GALV have a more restricted tropism and more specifically target hematopoietic stem cells. The provision of an efficient method for purifying pseudotyped vectors with GALV glycoproteins therefore represents a major challenge in the field of gene therapy.
[0002] The inventors have therefore proposed to develop a process for purifying enveloped viruses, and in particular viruses pseudotyped by the GaLV envelope glycoprotein or other envelope proteins, in order to produce virus preparations for clinical use.
[0003] SUMMARY OF THE INVENTION The present invention results from the unexpected observation made by the inventors of the influence of the pH of the solutions used during the purification of an enveloped virus, and of the positive influence of certain additives on the yield. of said purification. The present invention results in particular from the observation of the striking improvement in the purification of enveloped viruses when acidic buffers are used during anion exchange chromatography. The subject of the invention is therefore a process for purifying enveloped viruses comprising an anion exchange chromatography step, the buffers used during said chromatography being of pH less than 6. In particular, the pH may in particular be between , 5 and 6. According to one alternative, the pH of the buffers used during the anion exchange chromatography is greater than or equal to 6 and further comprises a polyol. The inventors have been able to show that the addition of a polyol in a or more than one of the buffers used in one or more steps of an enveloped virus purification process provided a substantial increase in purification efficiency. In particular, the improvement of the yield of a purification comprising an ultrafiltration / diafiltration step followed by anion exchange chromatography is observed when the buffers used during this chromatography comprise a polyol. The inventors also propose the inversion of the order of the anion exchange chromatography and ultrafiltration / diafiltration steps, in particular tangential flow filtration (TFF), used during the purification. The application of ultrafiltration / diafiltration, in particular of a TFF, before anion exchange chromatography allows a substantial improvement in the efficiency of the purification of an enveloped vector. The invention therefore also relates to a method for purifying an enveloped virus, comprising, in this order, an ultrafiltration / diafiltration step, in particular a TFF step, followed by an anion exchange chromatography step. The invention furthermore relates to a method for purifying enveloped viruses, said method comprising an ultrafiltration / diafiltration step, in particular a TFF step, said step being carried out using buffers containing a polyol.
[0004] The invention is more particularly adapted to the purification of pseudotyped viruses with a glycoprotein derived from GaLV. Until the provision of the present invention, the viruses pseutotyped with this type of glycoproteins were considered "non-purifiable".
[0005] The various studies carried out on these viral vectors used crude preparations of unpurified vectors because of the fragility of the pseudotyped GaLV vectors. Unexpectedly, the inventors have been able to show a significant improvement in purification efficiency by the method of the invention. They have also been able to show that this method also makes it possible to improve the efficiency of purification of pseudotyped viruses by means of other envelope glycoproteins, in particular by means of VSV-G and MV glycoproteins. DETAILED DESCRIPTION OF THE INVENTION Virus Production and Enveloped Vectors The production of enveloped viruses or vectors is well known in the state of the art. Those skilled in the art can refer to their general knowledge in this field, in particular represented by Ansorge et al. 2010; Schweizer and Merten 2010; Rodrigues et al. 2011. The virus produced is notably an enveloped viral vector. The viral vector is in particular derived from a retrovirus, in particular a lentivirus. The retroviral vectors produced are in particular derived from alpharetroviruses (such as VLA or ALV in English for avian leukosis virus), betaretroviruses (such as VTMM or MMTV in English for mouse mammary tumor virus), gammaretroviruses (such as different types of VLM or MLV in English for murine leukemia virus), deltaretrovirus (such as the different types of VLTH or HTLV in English for human T-lymphotropic virus), epsilonetrovirus (such as VSDW or WDSV in English for Walleye dermal sarcoma virus), spumavirus (such as VFH or HFV for human foamy virus and VFS or SFV in English for simian foamy virus), primate lentiviruses such as the different types of human immunodeficiency virus (HIV or HIV in English for human immunodeficiency virus), the different types of simian immunodeficiency virus (SIV), or non-primate mammalian lentiviruses such as simian immunodeficiency virus equine infectious anemia virus (EIAV), Feline Immunodeficiency Virus (FIV), the arthritis Caprine encephalitis (VAEC, or CAEV in English for caprine arthritis-encephalitis virus), or the ovine virus Visna-maëdi (VVM or VMV in English for visna maedi virus). According to one particular embodiment, the enveloped virus, in particular the retroviral vector, in particular lentiviral, is pseudotyped, that is to say that it comprises an envelope glycoprotein derived from a virus different from the virus from which it is derived. the retroviral particle, a modified envelope glycoprotein or a chimeric envelope glycoprotein. According to one particular embodiment, the retroviral vector is pseudotyped with an envelope glycoprotein derived from the vesicular stomatitis virus (VSV-G), the measles virus (MV for measles virus in English) or the gibbon leukemogenic virus. (GALV in English for gibbon ape leukemia virus), although one skilled in the art may consider the use of other viral envelope glycoproteins (Frecha et al., 2008). According to a particular embodiment, the enveloped virus, in particular the retroviral vector, more particularly lentiviral, is pseudotyped with a modified envelope glycoprotein such as GALVTR (GALV envelope glycoprotein whose C-terminus intravirion has been replaced by the C-terminal end of the envelope glycoprotein of amphotropic human leukemia virus A-MLV, thus permitting incorporation of the highly efficient envelope glycoprotein into the lentiviral particle) (Christodoulopoulos and Cannon 2001). According to a particular embodiment, the enveloped virus, in particular the retroviral vector, more particularly lentiviral, is pseudotyped with a chimeric envelope glycoprotein such as the envelope glycoprotein of the measles virus into which a fusion protein has been inserted. encoding the variable heavy and light chain variable immunoglobulin (scFv) region or an ankyrin repeat protein (DARPins) to allow specific targeting of a given receptor on the surface of target cells (Anliker et al., 2010, Münch et al., 2011). According to a particular embodiment, the lentiviral vector is pseudotyped with an envelope derived from the GALV virus, vesicular stomatitis virus (eg VSV-G envelope protein), or measles virus, although the man of the The profession may consider the use of other envelope proteins. The enveloped virus may further contain a transgene of interest introduced into its genome. Of course, the transgene of interest will depend on the specific use for which the enveloped viral vector is intended. Illustratively, let us mention a transgene of interest encoding a therapeutic RNA (eg a transgene of interest encoding an antisense RNA complementary to a target RNA or DNA sequence), a gene therapy transgene encoding a protein deficient or absent in a a subject suffering from a pathology, or a transgene used for a DNA vaccination, that is to say a transgene encoding a protein the expression of which will induce a vaccination of the recipient organism against said protein. According to one particular embodiment, an enveloped viral vector that can be used in gene therapy is produced and then purified. The production method used is advantageously compatible with good laboratory practices and makes it possible to envisage large-scale production of enveloped viral vectors, in particular retroviral vectors, especially lentiviral vectors, in particular pseudotyped lentiviral vectors (in particular with proteins GALV envelopes for a retrovirus or GALVTR for a lentivirus, VSV-G, or MV). According to a preferred embodiment for the production of a lentiviral vector, the following four elements are introduced into the host cell: an expression cassette comprising a lentiviral gagpol gene, an expression cassette comprising a lentiviral rev gene, a Expression cassette of a transgene of interest, between a 5'-LTR and a lentiviral LTR-3 ', and an envelope glycoprotein expression cassette (s). In a particular embodiment, the enveloped virus, in particular a retroviral vector, more particularly a lentiviral vector, is produced from a stable line expressing one or more elements necessary for the production of an enveloped virus (Miller 2001; Rodrigues et al., 2011), such as the human producer line GPRG-EF 1 ahy, OPT which constitutively produces a lentiviral vector derived from HIV-1 pseudotyped with the VSV-G envelope glycoprotein (Greene et al., 2012), or for example the murine PG13-MIG-GFP producing line which constitutively produces the MLV gammaretraviral vector pseudotyped with the GaLV envelope glycoprotein (Miller et al 1991). In a particular embodiment, the enveloped virus is produced from a mammalian host cell transiently transfected with one or more plasmids encoding the elements necessary for the production of the virus. According to a variant allowing the production of a lentiviral vector, said elements are introduced into the cell by means of 4 plasmids: a plasmid carrying an expression cassette comprising a lentiviral gagpol gene, a plasmid carrying an expression cassette comprising a gene reventiviral, a transfer plasmid comprising an expression cassette of a transgene of interest, between a 5'-LTR and a lentiviral LTR-3 'and a plasmid carrying a glycoprotein (s) expression cassette. 'envelope.
[0006] The host cell may be selected from any cell allowing the production of an enveloped virus. According to a particular embodiment, said cell is chosen from a human cell (HEK293, HEK293T, HEK293FT, Te671, HT1080, CEM), a murine cell (NIH-3T3), a mustelide cell (Mpf), a cell of canine (D17) (Miller and Chen 1996, Miller 2001, Merten 2004, Rodrigues et al 2011, Stacey and Merten 2011). The cells are cultured in a medium suitable for culturing mammalian cells and producing an enveloped virus. The medium may also be supplemented with additives well known in the art such as antibiotics, serum (including fetal calf serum, etc.) added in appropriate concentrations. The medium used may in particular comprise serum or be devoid of it. Mammalian cell culture media are well known in the art. Examples include DMEM (Dulbecco's Modified Eagle's medium), RPMI1640 or a mixture of different culture media, including, for example, DMEM / F12, or a serum-free medium such as optiMEMe, optiPRO ®, optiPRO-SFM®, CD293®, Freestyle F17® (Life Technologies) or Ex-Celle 293 (Sigma-Aldrich). In methods employing transiently transfected cells, any agent for transfection of plasmids can be used. It may be especially used calcium phosphate or polyethyleneimine, although other agents may be considered by the skilled person (Ansorge et al., 2010). The conditions (in particular amount of plasmid (s), ratio between the plasmids, ratio between the plasmid (s) and the transfection agent, the type of medium, etc.) and the transfection time may be adapted by those skilled in the art depending on the characteristics of the product virus and / or the transgene introduced into the transfer plasmid. The enveloped virus is then harvested from the culture supernatant according to methods well known in the art.
[0007] According to a particular embodiment, the culture medium used has a neutral pH (eg between 7 and 7.4, especially 7, 7.1, 7.2, 7.3 or 7.4) conventionally used in the state of the art for cell culture and virus production. According to one particular embodiment, the production method used comprises the cultivation of the producer cells in a moderately acidic medium. While the state of the art demonstrates the neutrality of culture media as a necessary condition for optimal cell culture and optimal production of enveloped viruses and vectors, it has been found that moderately acidic conditions on the contrary make it possible to improve significantly the production of an enveloped virus, in particular a lentivirus, in particular a pseudotyped lentivirus (for example with the GaLV protein (or GaLVTR), VSV-G, or envelope proteins of the measles virus ). According to one variant, the production of enveloped vectors is carried out in a moderately acidic condition. The term "mildly acidic condition" refers to the pH of an aqueous solution of between 5 and 6.8, especially 5.5 to 6.5, more preferably 5.8 to 6.2. According to a particular embodiment, the pH of the culture medium is about 6. The pH chosen will also depend on the buffering capacity of the culture medium used, which the skilled person can easily determine given his general knowledge. Those skilled in the art are able to modify the pH of a solution, in particular the pH of a cell culture medium. It may in particular introduce into said solution a solution of an acid, especially a strong acid such as hydrochloric acid. If necessary, a solution of a base, especially a strong base such as sodium hydroxide, can be used to readjust the pH upwards to reach the desired value.
[0008] According to a particular embodiment, the production of the enveloped virus comprises the following steps: transient transfection of HEK293T cells by means of one or more plasmids encoding the elements necessary for the production of said enveloped vector or the use of stable producer cells producing the vectors constitutively or after induction; culturing said cells in a suitable medium, whose pH is about 6 or about 7; harvesting the virus wrapped in the culture supernatant.
[0009] According to a variant of this embodiment, the enveloped virus produced is a lentivirus produced after transfection of the cells by means of four plasmids: a plasmid carrying an expression cassette comprising a lentiviral gagpol gene, a plasmid carrying an expression cassette comprising a lentiviral rev gene, a transfer plasmid comprising a transgene expression cassette of interest, between a 5'-LTR and a lentiviral LTR-3 'and a plasmid carrying a glycoprotein expression cassette (s). ) envelope. Alternatively, the coat protein is derived from the GaLV virus (particularly the modified glycoprotein GaLVTR for lentiviral vectors), VSV virus (in particular VSV-G envelope) or measles virus (MV).
[0010] Purification of viruses and enveloped vectors As shown in the examples of this application, the purification efficiency of the produced enveloped viruses can be strikingly improved by adapting the pH conditions of the buffers used during said purification.
[0011] In particular, the enveloped viruses can be very advantageously purified according to a new process comprising the use of acidic buffers during anion exchange chromatography. Thus, the invention relates in particular to a process for purifying an enveloped virus, the method comprising an anion exchange chromatography step, wherein the buffers used during this anion exchange chromatography step are of lower pH. or equal to 6.9. The enveloped virus may in particular be purified from the culture medium of a cell culture of cells producing said enveloped virus. In a particular embodiment, the pH of the buffer (s) used during the anion exchange chromatography step is between 4.5 and 6.2, more particularly between 5 and 6, in particular between 5.5 and 6, the pH being more particularly about 5.5 (eg 5.4, 5.5 or 5.6) or 6 (eg 5.9, 6 or 6.1), more particularly 5.5. The anion exchange chromatography may be preceded or followed, preferably preceded, by an ultrafiltration step, in particular an ultrafiltration / diafiltration, in particular a tangential flow filtration. According to one embodiment, anion exchange chromatography precedes ultrafiltration. According to an alternative method, ultrafiltration precedes anion exchange chromatography, the latter mode being preferred. The inventors have also shown the advantage of using a polyol, especially sucrose, in the buffers employed during anion exchange chromatography, when said buffers have a pH greater than or equal to 6, in particular a pH between 6 and 8. The invention therefore also relates to a process for purifying an enveloped virus comprising an ultrafiltration / diafiltration step followed by an anion exchange chromatography step, said chromatography being carried out using buffers of higher pH. or equal to 6, in particular of pH between 6 and 8, more particularly between 7 and 8, containing a polyol. The buffer (s) used during the ultrafiltration / diafiltration step, more particularly TFF, may also be acidic, neutral or basic buffers.
[0012] The person skilled in the art is able to modify the pH of a solution, in particular the pH of a buffer used during a purification stage of enveloped viruses. It may in particular introduce into said solution a solution of an acid, especially a strong acid such as hydrochloric acid, to reduce the pH or adjust it. If necessary, a solution of a base, especially a strong base such as sodium hydroxide, can be used to obtain a basic pH or readjust the pH upwards to reach a desired acid value. Of course, those skilled in the art will take care to use the appropriate formulation to obtain a buffer capacity solution adapted to the desired pH.
[0013] The solution loaded on or in the ultrafiltration / diafiltration device or on the anion exchange chromatography column may correspond to the cell culture supernatant optionally pretreated with benzonase and / or low speed centrifugation and / or a clarification. It is understood that this culture supernatant possibly pretreated does not correspond to a "purification buffer". However, its pH can also be adjusted before loading if needed. If the production has been carried out at neutral or acidic pH, the optionally pretreated culture supernatant can be loaded directly, or its pH can be decreased or increased before loading. It is also possible to envisage the addition of additives in the culture supernatant possibly pretreated before loading. For example, it is possible to add at this stage a polyol, an antioxidant (especially L-histidine, L-methionine, L-cysteine, glutathione or vitamin C), a metal salt, especially a magnesium salt such as MgCl 2 or MgSO4, or any other suitable additive. According to a particular embodiment, the culture supernatant, optionally pretreated, is loaded directly onto or in the ultrafiltration / diafiltration device or on the chromatography column, without pH adjustment and without addition of additive. Buffers of acidic, basic or neutral pH may be, and preferably are, used beforehand to balance the ultrafiltration / diafiltration device and / or to carry out ultrafiltration / difiltration or anion exchange chromatography as such. . According to one variant, the ultrafiltration / diafiltration step is a TFF step. In this variant, the diafiltration is carried out with a buffer whose pH is adjusted according to the modalities described above. Thus, the buffer used may be an acid buffer, in particular a buffer with a pH of less than 6 or a pH buffer of between 4.5 and 6.2, in particular between 5 and 6, in particular a buffer of pH 5.5 at pH 6, more particularly a buffer of pH 5.5. In a variant, the buffer used is a buffer of pH greater than or equal to 6, in particular between 6 and 8, more particularly between 7 and 8, comprising a polyol. The buffers used during the ultrafiltration step and during the diafiltration step may be different or identical. In a particular embodiment, the ultrafiltration step is carried out using a buffer having a pH of about 7 (in particular a pH of between 6.8 and 7.2, more particularly a buffer of pH 7). and the diafiltration is carried out by means of a buffer having a pH of between 4.5 and 6.2, in particular a pH of between 5 and 6, more particularly a pH buffer of between 5.5 and 6, in particular a pH buffer equal to 5.5.
[0014] In one embodiment, the purification process comprises an anion exchange chromatography step followed by an ultrafiltration / diafiltration step. In another embodiment, the purification method comprises an ultrafiltration step followed by an anion exchange chromatography step.
[0015] In a particular embodiment, the purification method comprises: (a) clarifying the cell culture medium; (b) an ultrafiltration / diafiltration step; (c) anion exchange chromatography; (d) exclusion chromatography; steps (b) and (c) can be reversed. According to a preferred embodiment, step (c) follows step (b). Figure 1 of this application summarizes the steps of a preferred embodiment of the purification process of the invention.
[0016] According to a particular embodiment, a first clarification step is carried out by filtration of the culture supernatant on a filter whose retention point is between 0.2 and 0.8 μm, in particular a 0.45 μm filter, and recovering viruses wrapped in the filtrate. According to one embodiment, the clarification is performed by means of a cascade of different retention threshold filters, for example with a succession of filters 0.8, 0.45 and 0.21.1m or else 0.65 and 0. This clarification step may be preceded by a centrifugation step of the culture supernatant at low speed. The centrifugation speed at this stage may especially be between 500 g and 1000 g.
[0017] According to a particular embodiment, the ultrafiltration / diafiltration step is in particular carried out by tangential flow filtration. According to this embodiment, the tangential flow filtration can be carried out by means of a hollow fiber membrane or a flat membrane cassette or a spiral wound membrane (or "spiral wound membrane", in English), whose size the pore exclusion is between 300 and 800 kDa, in particular between 500 and 750 kDa. In another embodiment, the pore exclusion size is at least 300, 400, 500, 600, or 700 kDa, or 800 kDa. In a preferred embodiment, the membrane used for tangential flow filtration is characterized by a pore exclusion size of 750 kDa. According to a particular embodiment, at the end of the ultrafiltration / diafiltration step, the enveloped viruses, in particular the enveloped viruses present in the filtrate of the clarification step, are concentrated to the minimum possible volume, for example at least 5X, at least 10X, 15X, 20X, 25X, 30X, 35X, 40X, 45X, 50X, 55X or 60X. For example, the enveloped viruses are concentrated at 36.6X or 50X at the end of the ultrafiltration / diafiltration step. According to another embodiment, the ultrafiltration / diafiltration step makes it possible to reduce the contaminant load by more than 70%, 80%, 85%, 90% or more than 95%. According to a particular embodiment, the ultrafiltration / diafiltration step is carried out in the following manner: a first concentration is carried out, for example a concentration of 25 × (in particular, passing from a volume of 500 ml to a volume of 20 ml), followed by diafiltration with at least 2, 3, 4, 5, 6, 7, 8, 9, or even at least 10 volumes of an acidic buffer containing or not a polyol and / or one or more antioxidants for example with 10 volumes of buffer, this step is then followed by a second concentration step, in particular to the minimum possible volume, to reach for example a concentration of 50 X.
[0018] Anion exchange chromatography supports are well known in the art. The invention more particularly uses anion exchange chromatography on a column or membrane, more particularly on a column. A preferred embodiment comprises the use of weak anion exchange chromatography (in particular DEAE (D) -diethylaminoethyl, PI-polyethylenimine). As such, there may be mentioned the use of a support selected from a DEAE column. As an illustration, mention may be made of the supports Monolith CIM D (BlAseparation), Poros D50 (Life Technologies), Sartobind D (Sartorius), Toyopearl 650C DEAE (Tosoh), etc. In a preferred variant, the anionic exchange chromatography support is a non-compressible support such as the supports Monolith CIM D and Poros D50, allowing to obtain a better yield than the compressible supports. When the pH of the buffers used during the anion exchange chromatography is less than 6, said buffers contain or do not contain a polyol. In particular, it has been observed that the use of buffers of pH 5.5 during this step made it possible to obtain a yield of approximately 100% of the pseudotyped lentiviral vectors GALVTR without the necessity of adding a polyol. This excellent level of yield has never been achieved so far and makes it possible to envisage the large-scale use of this type of particularly advantageous vectors for the reasons set out above. The addition of a polyol has not shown any positive or negative impact on this yield, but their addition may be considered for the purpose of stabilizing the eluted vectors. When the pH of the buffers used is greater than or equal to 6, in particular between 6 and 8, the addition of a polyol significantly improves the purification yield. Thus, according to the invention, when the pH of the anion exchange chromatography buffers is between about 6 and about 8, said pHs contain a polyol. In one embodiment, the purified and filtered solution of the ultrafiltration / diafiltration step is deposited on the chromatography medium which was first equilibrated by means of a pH equilibration buffer of less than 6 containing optionally a polyol, especially a 20 mM Bis-Tris buffer, pH 5 to 6 (for example pH 5.5), 5% sucrose, 2 mM MgCl 2 or a pH equilibration buffer greater than or equal to 6, in particular understood between 6 and 8, containing a polyol, for example a PB S buffer, pH 7, 5% sucrose or Bis-Tris-propane pH 8, 5% sucrose, 2 mM MgCl 2. The buffer is introduced at a suitable rate (for example at 1 column volume / min or 4 cm / min). Then the column is washed with the equilibration buffer, and finally eluted. According to one particular embodiment, the elution of the anion-exchange chromatography column is carried out in two stages with a buffer that may or may not comprise a polyol, in particular with a 0.3M NaCl, 20 mM Bis-Tris buffer (pH 5 to 6, in particular pH 5.5), 5% sucrose, 2 mM MgCl 2 and then the elution of the vectors is carried out with an upper ionic strength buffer comprising a polyol, in particular a 650 mM NaCl buffer, 20 mM Bis-Tris ( pH 6), 5% sucrose, 2 mM MgCl 2. The method according to the invention therefore advantageously makes it possible to reduce the amount of salts necessary to elute the vectors of the column relative to the concentration of salts conventionally used. For the evaluation of the fractions and the selection of those which will be submitted as a result of the purification process, the column may be equipped at the output of a chromatograph equipped with a 280 nm UV absorbance reader, a conductivity meter, a plotter and a fraction collector. An exclusion chromatography step is preferably carried out immediately after the anion exchange chromatography step. The exclusion resin used has an exclusion size of between 300 and 1000 kDa, in particular between 500 and 800 kDa. According to a particular embodiment, the exclusion chromatography column used is defined by an exclusion size of 500, 700 or 800 kDa. Moreover, in a particular embodiment, the column used for this step comprises a multimodal resin, in particular a resin with a double functionality of exclusion and adsorption gel (by hydrophobic interaction as well as by a positive charge of the support). One can cite the Capto Core700 column (GE HealthCare). In one embodiment, the chromatography gel is a non-compressible support. The sample of viruses or enveloped vectors from anion exchange chromatography is loaded onto the column and then the formulation buffer is injected at a suitable rate, for example at a rate of 0.5 ml / minute. The fractions to be collected are determined at the column outlet by means of a UV absorbance reader at 280 nm. According to one aspect, the invention relates to a process for purifying enveloped viruses comprising an ultrafiltration / diafiltration step, in particular a TFF, said step being carried out using buffers containing a polyol. The inventors have been able to show that the addition of a polyol in an ultrafiltration / diafiltration buffer allows a significant increase in the purification yield (Table 2 below, in the examples), which had never been reported. The polyols that can be used during this step and their concentration are specified below. The buffers used during the ultrafiltration step and during the diafiltration step may be different or identical. In a particular embodiment, the ultrafiltration step is carried out using a buffer having a pH of about 7 (in particular a pH of between 6.8 and 7.2, more particularly a buffer of pH 7). and the diafiltration is carried out by means of a buffer having a pH of less than 6, or of between 4.5 and 6.2, in particular a pH of between 5 and 6, more particularly a pH buffer of between 5.5 and 6, more particularly a pH buffer equal to 5.5 or 6, in particular a pH buffer equal to 5.5. Tangential flow filtration may be carried out by means of a hollow fiber membrane or a flat membrane cassette or a spiral wound membrane (or 'spiral wound membrane'), the pore exclusion size of which is between 300 and 800 kDa, in particular between 500 and 750 kDa. In another embodiment, the pore exclusion size is at least 300, 400, 500, 600, or 700 kDa, or 800 kDa. In a preferred embodiment, the membrane used for tangential flow filtration is characterized by a pore exclusion size of 750 kDa. According to a particular embodiment, at the end of the ultrafiltration / diafiltration step, the enveloped viruses, in particular the enveloped viruses present in the filtrate of a clarification step, are concentrated to the minimum possible volume, for example at least 5X, at least 10X, 15X, 20X, 25X, 30X, 35X, 40X, 45X, 50X, 55X or 60X. For example, the enveloped viruses are concentrated at 36.6X or 50X at the end of the ultrafiltration / diafiltration step. According to another embodiment, the ultrafiltration / diafiltration step makes it possible to reduce the contaminant load by more than 70%, 80%, 85%, 90% or more than 95%. According to a particular embodiment, the ultrafiltration / diafiltration step is carried out in the following manner: a first concentration is carried out, for example a concentration of 25 × (in particular, passing from a volume of 500 ml to a volume of 20 ml), followed by diafiltration with at least 2, 3, 4, 5, 6, 7, 8, 9, or even at least 10 volumes of an acidic buffer containing or not a polyol and / or one or more antioxidants , for example with 10 volumes of buffer, this step is then followed by a second concentration step, in particular to the minimum possible volume, to reach for example a concentration of 50 X. The ultrafiltration / diafiltration step may be followed or preceded by an anion exchange chromatography step, or followed by an exclusion chromatography step, according to the modalities described in detail above. According to a particular embodiment, the purification process is intended for production of enveloped viruses of research grade. In this case, the ion exchange chromatography step may be omitted (the method then comprises for example the steps (a), (b) and (d) described above). According to another embodiment, the method is intended for the purification of clinical grades of virus. In the latter case, the anion exchange chromatography step is preferably included. Furthermore, the exclusion chromatography step may be followed by a sterilizing filtration step, in particular by means of a filtering membrane, in particular a retention threshold membrane less than or equal to 0.22 nm. In the context of the present invention, the term "polyol" defines a linear, cyclic or bicyclic carbon molecule comprising between 3 and 18 carbon atoms, in particular between 3 and 12 carbon atoms, substituted by at least 3-6 hydroxyl groups. , in particular 8 hydroxyl groups. The polyol may for example be a monosaccharide aldose or ketose, in particular a tetrapose, a pentose or a hexose. Mention may in particular be made of the following monosaccharide polyols: erythrose, threose, ribose, arabinose, xylose, lyxose, allose, altrose, glucose, mannose, gulose, idose, galactose, talose, erythrulose, ribulose, xylulose, fructose, psicose, sorbose, tagatose. . The polyol may also be chosen from the following disaccharides or trisaccharides which constitute a nonlimiting list of other polyols that may be used in the practice of the invention: cellobiose, gentiobiose, inulobiose, isomaltose, isomaltulose, kojibiose, lactose, lactulose, laminaribiose , leucrosis, maltose, maltulose, melibiose, nigerosis, robinosis, rutinose, sucrose, sophorosis, trehalose, trehalulose, turanose, erlose, fucosyllactose, gentianosis, inulotriosis, 1-kestosis, 6-kestosis, maltotriosis, mannotriosis, melezitosis, neokestosis, panosis , raffinose, rhamninose. In a particular embodiment, the polyol is chosen from raffinose, isomaltotriose, sucrose, mannitol, sorbitol, trehalose, glucose and glycerol. In a particular embodiment, the polyol is sucrose.
[0019] The concentration of polyol may vary to a large extent, and may in particular be different for each of the different buffers implemented during this step. The concentration of polyol may especially be between 1% and 15% (w / v) especially between 1.5% and 10%, in particular between 2% and 8%, more particularly between 2% and 5%. In a particular embodiment, the polyol concentration of one or more of the buffers of the anion exchange chromatography is 5% (w / v). In a particular embodiment, all the buffers used during the purification process comprise a polyol, in particular sucrose, in particular at 5% (w / v). Thus, according to this embodiment the method may comprise a TFF step, an anion exchange chromatography step and an exclusion chromatography step in which all the buffers used comprise a polyol. In another embodiment, the buffers of the TFF and anion exchange chromatography steps comprise a polyol while the buffers used to equilibrate and elute the exclusion chromatography column do not include a polyol, these buffers corresponding to the buffer. formulation whose composition will largely depend on the therapeutic purpose and the mode of administration of the finished product. In another embodiment, one or more of the buffers used in the process of the present invention, including buffers used during ultrafiltration / diafiltration and / or anion exchange chromatography, include a magnesium salt. , especially magnesium chloride or magnesium sulfate. The concentration of magnesium salt, in particular magnesium chloride or magnesium sulfate in each of the buffers can, independently for each buffer, be between 0.1 mM and 5 mM, especially between 1 and 3 mM, in particular 2 mM.
[0020] In another embodiment, one or more of the buffers used in the process of the present invention include L-His, L-Met, L-Cys, glutathione, or vitamin C for inactivating free radicals. The concentration of these components in each of the buffers may, independently for each buffer, be between 0.1 mM and 20 mM.
[0021] The purification process according to the invention may also comprise one or more stages of treatment of the sample or samples with a nuclease, in particular a benzonase. Nuclease may be used before or after each step. In one embodiment, nuclease, in particular benzonase, is used in the culture medium of the producer cells after the transfection step of the plasmids. According to one embodiment, one or more purification stage (s) are carried out at a temperature below room temperature, in particular at a temperature of between 2 and 12 ° C., more particularly between 4 and 10 ° C. According to a particular embodiment, one, more or all the steps of the purification are carried out at about 4 ° C. LEGEND OF FIGURES FIG. 1. Two processes for the purification of LV-GaLV-TR lentiviral particles: process (A) is a simplified process employing a single exclusion chromatography step (gel filtration) after the step of tangential filtration in flux; and process (B) is a more elaborate process for obtaining a higher purity than when using process (A) - for example for the production of vectors for clinical use.
[0022] Fig. 2. Comparison of membranes with a cut of 500 kDa and 750 kDa in the removal of contaminating proteins (SDS-PAGE (top) and Western Blot anti-p24 (bottom)). The band at 24/25 kDa (= p24) is clearly visible for all samples on the Western Blot: 1) size markers; 2) diafiltration (a) (test 1) at 750 kDa; 3) diafiltration (b) (test 2) at 750 kDa; 4) ultracentrifugation at 68338g for 3h (resuspension in X-vivo culture medium 20); 5) diafiltration (a) (test 1) at 500 kDa; 6) diafiltration (b) (test 2) at 500 kDa; 7) culture supernatant containing LV-GaLV-TR vectors. Fig. 3. Effect of NaC1 on the stability of LV-GaLV-TR vectors encoding GFP stored at room temperature. The vectors are incubated for 4 hours at pH 7.0 (PBS) at room temperature (RT). To optimize the anion exchange chromatography step, we tested the stability of the vectors in a saline NaCl medium. For this, the vectors were incubated after the UF / DF step in pH 7.0 PBS buffers of different NaC1 concentrations for 4 hours at room temperature. Then the vectors were titrated on HCT116 cells. 48 hours later, cells were switched to the FACS to measure the percentage of expression of GFP.
[0023] Fig. 4. Effect of pH and salinity of the elution buffer on the purification efficiency of the infectious lentiviral vectors after an anion exchange chromatography step. The vector preparation was produced by transfection of HEK293T cells, clarified and concentrated / Diafiltered by TFF for use in the evaluation of different (weak) anion exchange media. Different supports were evaluated: Toyopearl 650C DEAE, CIM D (DEAE) and Poros D. The 100% yield is equivalent to the infectious titre after the previous step of TFF. Fig. 5. Purification of a lentiviral vector preparation GaLV-TR by exclusion chromatography (Capto Core 700). Three ml of a lentiviral preparation was concentrated / diafiltered and then passed through a column of Capto Core 700 (4.5m1). PBS buffer (pH 7.0), 5% sucrose, 2mM MgCl 2 was used during this step for column equilibration and formulation. Fractions of 1 ml were collected and analyzed for the vector concentration (TU): a) Chromatogram showing the titer (TU) by fraction, the cumulative amount of vector for fractions 4-9 and cumulative recovery (%) for fractions 4-9; b) Western blot of all fractions; c) SDS-PAGE of all fractions. Fig. 6. Transduction of CD34 + SC (ambilical cord blood) cells with HIV-GaLV-TR MOI vector. Gross: percentage of cells expressing GFP determined by flow cytometry of CD34 + cells transduced with the HIV-GaLV-raw product. TR, DF / UF: percentage of cells expressing GFP determined by flow cytometry of CD34 + cells transduced with the preparation of HIV-GaLV-TR vectors obtained after purification and concentration by TFF of the crude product.
[0024] EXAMPLES The work reported in this application was funded through the 7th Framework Program of the European Community (FP7 / 2007-2013) under number 222878. Materials and methods Cells: HEK293T and HCT116 cell lines (Colorectal cancer cells CCL-247, origin: ATCC) are cultured at 37 ° C, 5% CO2 in Dulbecco's Modified Eagle's medium (Gibco) (DMEM + Glutamax) supplemented with 2-10% fetal calf serum (FCS) (Life Technologies). Culture medium: DMEM / SVF buffered to pH 6.0 by the addition of hydrochloric acid (37% HC1, Sigma-Aldrich), then filtered using a Corning® 1000mL filter (0.221 μm PES (polyethersulfone)).
[0025] Viral vector production: Virus vectors derived from HIV-1 pseudotyped with different glycoproteins are produced by transient calcium phosphate transient transfection in 293T cells, by 4 plasmids as described by Merten et al. (2011). 2x108 293T cells are seeded in Hyperflask 1760-cm2 (Corning) in 550m1 of DMEM 10% FCS (Kutner et al., 2009). 24 hours later, the culture medium is replaced by the transfection medium, combining the DNA / CaCl 2 / HBS complex in it. Plasmids: gagpol (pKLgagpol) 1361.1g, rev (pKrev) 52.251.1g, transgene plasmid (pCCL-eGFP) 206.81.1g, envelope plasmid suitable for each pseudotype: GaLV-TR: pBA.GALV-TR / AmphoKana (Gibbon ape Leukemia Virus) 2231.1g to generate LV-GaLV-TR; VSV-GpMDG (Vesicular stomatitis virus-g) 68.131.1g to generate LV-VSV-g; pFA30 and pHCMH2 (modified envelope proteins of measles virus) 401.1g and 141.1g to generate LVMV; sufficient quantity for 18m1 H20 and 8.9m1 TE0.1X, mix with 3m1 of CaCl2 (2.5M) then add 30m1 of HB S2X, wait for the formation of the complex for 4 min and add the mix to the culture medium. After 16 hours the supernatant is replaced by the fresh medium 2% SVF 15U Benzonase (Merck) and 2mM MgC12 (Sigma-Aldrich). The harvest is done after 48 hours post transfection the supernatant is filtered through a 0.451.1m cellulose acetate (CA) 1L filter (Corning). The MLV-GaLV retroviral vectors are produced by PG13 cells. These are cells producing the MLV-GaLV vectors (Miller et al., 1991), the cells are maintained in Dulbecco's modified Eagle's medium (Gibco) (DMEM + Glutamax) supplemented with 2 to 10% FCS, 37 ° C, 5% CO2. The harvest of the vectors is done after 24 hours of change of culture medium. Then the production supernatant is clarified on a 0.451.1m cellulose acetate filter (Corning).
[0026] Concentration of Viral Vectors by Tangential Flow Filtration (TFF): This step consists in concentrating the production supernatant and then replacing the culture medium with an appropriate buffer for the rest of the process. Ultrafiltration (UF) is performed after preparation of the UF cassette and determination of the standardized NWP water permeability at 0.5 bar at 20 ° C. The membrane is then equilibrated with Bis-Tris buffer pH 6.0 5% sucrose, 2mM MgC12 or other buffers to perform this concentration / diafiltration at other pHs (eg PB S, pH 7.0, 5% M, sucrose, 2mM MgCl 2). The entire process is carried out at about 4 ° C and the product reservoir to concentrate put in an ice bucket.
[0027] Principle: A first concentration to the volume of 20 ml followed by diafiltration with 10 volumes of ion exchange chromatography loading buffer (in this case: 10 x 20m1) are carried out. These steps are followed by a second concentration to the minimum possible volume (10 ml in this case).
[0028] Membrane 750 kDa, 410 cm2: "Hollow fiber cartridge" (GE Healthcare, Ref: UFP750-E3MA) using the Kros-Flow research II TFF system (Spectrum). After validation of the integrity of the membrane, the concentration of the vectors begins with a starting volume of 500 ml of crude supernatant and is concentrated by the membrane of 500 ml to 20 ml.
[0029] The concentrated product is diafiltered against 200 ml of buffer A (to diafilter 10 times 20 ml of concentrate). This represents a concentration factor of 25X. The final volume of the diafiltrate is 10m1. In this case the concentration factor is 50X.
[0030] Anion exchange chromatography: In a first protocol, the anion exchange chromatography step is carried out downstream of the TFF. Several chromatographic supports are tested: CIMD DEAE monolithic column, CIMD Q (BlAseparation, Villach, Austria), column volume: 1m1; Sartobind D 75MA, volume: 2.1m1 (Sartorius Stedim Biotech); Poros PI, volume of the column: 4m1; Poros D 50, Column Volume: 4m1, Poros HQ, Column Volume: 4m1 (LifeTechnologies), Toyopearl 650C DEAE, Column Volume: 2m1 (Tosoh). The column to be tested is connected to a Biologic-LP (Biorad) chromatograph equipped with a 280 UV absorbance reader, a conductivity meter, a plotter (Chart recorder 1327, Bio-Rad), and a collector. Fraction (Mode! 2110, Bio-Rad).
[0031] The column is equilibrated with 5 column volumes (5 HP) of buffer A at 2m1 / min. After loading the sample onto the column, the column is washed with 5 CV of appropriate equilibration buffer according to the desired pH, according to Table A). Elution in two steps is then carried out: 0.3M NaCl, 20mM Bis-Tris, 5% sucrose, 2mM MgCl2 (pH 6.0) then 650mM NaCl, 20mM Bis-Tris, 5% sucrose, 2mM MgCl2 (pH 6, 0) to elute the vectors.
[0032] Three other pH's are tested pH 5.5, 7.0 and 8.0 using the appropriate buffers (including buffers such as Bis-propane, PBS, L-His); in the presence (5%) and in the absence of sucrose 5% and MgCl 2 (2 mM). Finally, the fraction is loaded immediately on the gel filtration column (exclusion chromatography) to remove the contaminants and contaminant proteins eluted with the 650 mM NaCl vectors. In a second protocol, the clarified production supernatant is loaded onto a Poros D anion exchange chromatography column, without a prior ultrafiltration / diafiltration step to evaluate the chromatography yield under these conditions. The equilibration buffer used has a pH of 5.5, and contains 5% sucrose and 2 mM MgCl 2. Exclusion Chromatography: This is the final step before sterilizing filtration for Methods A and B (Figure 1). This step consists in eliminating contaminants having a smaller size than the gel used (exp: 750kDa, or 500kDa). The Captocore700 column was used for this step. It is a gel with dual functionality: exclusion chromatography and adsorption chromatography gel. Before starting the loading, the column is sanitized with 1M NaOH and equilibrated with the formulation buffer. The product UF / DF (Method A), or the fractions corresponding to the peak of chromatography of anion exchange chromatography (AXC) (Method B) is loaded onto the column. 8 ml (UF / DF, or AXC fraction) are loaded at a flow rate of 0.5 ml / min.
[0033] Then the formulation buffer is injected at 0.5 ml / min (5 CV formulation buffer). The fraction corresponding to the UV peak is collected (approximately 16 ml) and then filtered on 0.221.1m filter (sterilizing filtration). Samples are stored at -80 ° C. Titration of viral vectors: The viral titer in transducing units (TU) of the vectors having the eGFP reporter gene is analyzed by transduction of HCT116 cells. At 72 hours post transduction cells were switched to FACS to determine the TU / ml titre as previously described (Pfeifer et al., 2009). For physical analysis of the virus particles, the p24 ELISA KIT (PerkinElmer) was used for the quantification of the p24 lentivirus capsid protein according to the supplier's instructions. Transduction of CD34 + umbilical cord blood cells: CD34 + cells are isolated from umbilical cord blood by immunomagnetic selection (Miltenyi Biotec). Culture and transduction of CD34 + cells is as described (Charrier et al., 2011): first, the cells are prestimulated overnight in X-Vivo (Lonza) medium and supplemented with cytokines. The pre-activated cells are inoculated in a 48-well plate (5'4 cells / 1041). The transduction is done by the addition of 1041 of vectors (1e6TU) purified in the presence of 8 μg / ml of vectofusin-1 (Fenard et al., 2013). After 6 hours of incubation, 1 ml of differentiation medium (X-VIVO-20 supplemented with 10% serum, and in the presence of cytokines (hSCF, h-II-3 hFlt3 h-Il-6) are added as described (Charrier et al. 2011)) in each well, and after 5 days the transduction efficiency is evaluated by measuring the expression of GFP by FACS (FC500, BD Biosciences).
[0034] Western blot SDS-page: The culture samples containing the lentiviral vectors or the purified samples are analyzed by SDS-PAGE and by Western Blot to detect the presence of the p24 capsid proteins. The p24 proteins are revealed according to the method developed by LI-COR, with the Odyssey device and the Odyssey 2.1 software. The primary antibody used is an anti-p24 (Santa Cruz # SC-57823) for the detection of HIV p24 capsid proteins. The antibody is used with a dilution of 1 / 200th in PBS1X-Tween 0.1% + Odyssey blocker (1: 1). The secondary goat antibody used coupled to Li-COR's "Dey 800" fluorochrome is directed against the primary antibodies.
[0035] Quantification of Residual Proteins and Specific Residual DNA: Total proteins are quantified by Bradford method (Bio-Rad) with serum albumin as standard. The test is done according to the instructions of the supplier. Residual DNA: The quantification of residual DNA of plasmid origin and / or from the host cell is done by quantitative PCR. The samples are treated with proteinase K (Roche) and the DNA is extracted using the MagNA Pure DNA and viral NA small volume kit system (MagNA Pure 96 Roche). Real-time quantitative PCR is then performed, with specific primers for the kanamycin gene to detect residual DNA of plasmid origin. To detect the residual DNA of the host cell, primers that target the E1A gene are used. Absolute quantification is performed with respect to a reference plasmid containing the amplified regions by quantitative PCR and whose copy number is known. Table A: Buffers used in the process Buffers pH Sucrose MgC12 (Sigma-Aldrich) (Sigma-Aldrich) Buffer A L-Histidine 20mM (Sigma-Aldrich) 5.0% W / V 2mM Buffer B Bis-Tris 20mM ( Sigma-Aldrich) 5.5 20mM Bis-Tris C buffer (Sigma-Aldrich) 6 PBS D buffer (GIBCO®) 7.2 20mM Bis-Propane E buffer (Sigma-Aldrich) 8 Results This invention relates to the development and establishment of a new protocol for purification of lentiviral vectors derived from HIV-1 or other retroviruses produced by transient transfection or with stable and pseudotyped producer cells with different envelope glycoproteins, such as GaLV-TR, VSV-G, measles virus and GaLV γ-retroviral vectors produced from stable cells, such as PG13, ensuring good yield and quality of purified virus particles. This development is essentially, but not exclusively, based on three purification techniques: TFF (tangential flow filtration), anion exchange chromatography and exclusion chromatography. The various combinations are illustrated in FIG. 1 Cell culture and clarification: These steps consist in producing retroviral and lentiviral vectors using stable cells such as PG13 characterized by stable and continuous production of retroviral vectors in continuous culture with a exchange of regular medium or cells such as HEK293 or HEK293T which must be transfected with 3 or 4 plasmids (providing the `helper 'functions of the lentivirus and the recombinant vector sequence) in order to induce the production of lentiviral vectors . The transient production is limited in time and allows one or more harvests a few days after transfection. Titers in general depend on the construction (sequence) of the vector but also the envelope protein. The following titles can be obtained with these production systems (Table 1). Table 1. Vector concentrations obtained with the different production systems: Production cell, vector concentration (TU / ml, g / ml) References vector-pseudotype PG13, MLV-GaLV-TR 5e6 TU / ml Miller et al. 1991 HEK293T, LV (HIV-1) -5e5 TU / ml Sakuma et al. 2010 GaLV-TR HEK293T, LV (HIV-1) -VSVg 1-5'7 U / ml Merten et al. 2011 Before any subsequent treatment it is possible to remove cell debris and aggregates present in the production supernatant. Conventionally, a filter of 0.45 μm (cellulose acetate) is used. The yield of this step is 80 ± 5%. However, those skilled in the art may use other membranes or membrane cascades, characterized by similar behavior and performance. Tangential flow filtration: Tangential flow filtration comprises two successive stages of ultrafiltration and diafiltration (UF / DF). These two steps make it possible to eliminate a large portion of the contaminants whose size is smaller than the pore exclusion size of the membrane used. This UF / DF stage also makes it possible to concentrate the viral particles and to reduce the volume of the product to be purified. A 110 cm 2 membrane with a pore exclusion size of 750 kDa (GE HealthCare) was used. Before starting the UF, different concentrations of sucrose (especially 5% sucrose (weight / volume)), and different concentrations of MgC12 (especially 2mM MgCl2 (final concentration)) are added to the clarified product. Then the concentration step by UF is at a flow of 80m1 / min, 7psig. The tank of the TFF is placed in an ice bucket to ensure a low temperature during the UF / DF. The diafiltration step begins after reducing the volume from 500m1 to 20m1 during the UF. For the DF, 200 ml (10 volumes of the concentrated product) of the diafiltration buffer are used: PBS, 5% sucrose, 2 mM MgCl 2. At the end of this step 20m1 of UF / DF product is recovered in a Corning 50m1 tube. The choice of buffer depends on the use of the preparation or the optimal conditions of the step following the concentration / diafiltration (eg in this case other buffers may be used such as Bis-Tris (pH 6, 0), 5% 2mM sucrose MgCl2) - see Table A. The samples are titrated on HCT116 cells as described by Fenard et al. (2013). Optimization studies of the concentration / diafiltration conditions: 1. The lentiviral particles have a diameter ranging from 80 to 120 nm, meaning that the pore size of membranes that can be used for concentration / diafiltration can go to max. up to about 50 nm (or 750 kDa). Within the scope of this invention, the 500 kDa and 750 kDa cut-off sizes have been evaluated. Yields (in UT) were as follows: 64% for the 750kDa membrane versus 34% TU yield for the 500kDa membrane.
[0036] Figure 2 shows the electrophoresis gels (SDS-PAGE and Western Blot) for vector preparations after tangential filtration using membranes with a cutoff of 500 kDa and 750 kDa. In addition to the higher yields obtained when using 750 kDa membranes, it is clear that a 750 kDa cutoff had a positive effect (Fig. 2, columns 2, 3) compared with the use of the 500 kDa membrane at the level of contaminant removal (Fig. 2, columns 5, 6). In addition, the concentrate generated with the 750 kDa membrane contains much less intense protein bands than observed for the crude supernatant. 2. Since the tangential filtration step is characterized by the generation of shear fields leading to the inactivation of the retroviral / lentiviral particles, it was necessary to optimize this step in order to maintain the functionality of these vectors. The addition of a polyol at different concentrations was evaluated in order to protect the lentiviral vector from the adverse conditions of the tangential filtration.
[0037] Table 2. Concentration / diafiltration yield of LV-GaLV-TR using different concentrations of sucrose. Yield% (TU) 0% sucrose 50.83 2% sucrose 80.31 5% sucrose 80.40 10% sucrose 52.37 15% sucrose 68.49 Note: 190 ml of crude supernatant was concentrated to 17 ml and several diafiltered with PBS (pH 7) + different% sucrose and 2mM MgCl 2.
[0038] These results clearly show the advantage of carrying out the concentration / diafiltration of supernatant containing LV-GaLV-TR vectors in the presence of sucrose and MgCl 2. The best yields are obtained at concentrations of 2% to 5% sucrose (Table 2). In addition, the use of a moderate concentration of sucrose has the advantage that the sample to be concentrated is less viscous because high concentrations of sucrose (10% - 15%) lead to an increase in viscosity. 3. Evaluation of pH and its Effect on Tangential Filtration and Functional Vector Efficiency: In the application FR 13 58909 filed by the present applicant, it has been shown that the production of enveloped vectors pseudotyped with different envelope proteins is increased when using pH 6.0 (up to 2x). It was decided to evaluate the impact of the choice of the pH of the supernatant containing the lentiviral vectors on the efficiency of the tangential filtration. In this context, two different pH values were evaluated (pH 6 and pH 7) during the concentration / diafiltration of GaLV-TR pseudotyped lentiviral vectors (Table 3). The reduction in pH from 7.0 to 6.0 resulted in a reduction in yield of about 10% (from 73.6% to 64%). However, this yield remains acceptable and it is therefore possible to envisage a concentration / diafiltration at acidic pH. Table 3. Impact of the pH of the supernatant to concentrate / diafiltration buffer on the concentration / diafiltration yields of pseudotyped lentiviral vectors GaLV-TR and VSV-g. Tangential filtration condition Vector LV Yield (%, TU) PBS, 5% sucrose 2mM MgC12, pH 7.0 LV-GaLV-TR 73.64 Bistris 20Mm, 5% sucrose, 2mM MgC12, pH 6.0 LV-GaLV-TR 63.99 4. Identification of the best condition for the concentration / diafiltration of lentiviral vectors GaLV-TR: For lentivirus GaLV-TR, the best concentration / diafiltration condition (tangential filtration) was as follows: LV-GaLV vectors - TR (1L) are clarified through a 0.45 μm cellulose acetate membrane, in the presence of 5% sucrose and 2mM MgCl 2, followed by the TFF step (750kDa cartridge, 410cm 2) with reduction of the volume to reach 20m 1 ( 50X). A diafiltration step is then performed against a volume of 200m1 of appropriate buffer (for example: 20mM Bis-Tris pH 6.0, 5% sucrose and 2mM MgCl2 or PBS pH 7.0, 5% sucrose and 2mM MgCl2).
[0039] The yield of this step for the LV-GaLV-TR vectors is 86% ± 5%, for a starting volume of 550m1 of the crude product. The volume of the concentrated product is 15m1 with a concentration factor of 36.6X and the elimination of contaminants reaches more than 90%. 5. Evaluation of established tangential filtration conditions for the concentration / diafiltration of other retroviral and lentiviral vectors pseudotyped with different envelope proteins: In the scientific literature different envelope proteins have been evaluated with a view to studying and improving the tropism retroviral and lentiviral vectors. In this context, the conditions established for the concentration / diafiltration of the lentiviral vectors GaLV-TR were evaluated for the concentration / diafiltration of retroviral and lentiviral vectors pseudotyped with different envelope proteins (Table 4). The results obtained with pseudotyped lentiviral vectors GaLV-TR are indicated as reference.
[0040] Table 4. Concentration / diafiltration of pseudotyped retroviral vectors with different envelope proteins: Tangential filtration condition Retroviral vector Yield% (TU) BISTRIS, 5% sucrose, 2mM MgC12, pH 6.0 MLV-GaLV (PG13) 94.2 PBS, 5% sucrose, 2mM MgC12, pH 7.0 LV-GaLV-TR 73.64 BISTRIS, 5% sucrose, 2mM MgCl2, pH 6.0 LV-GaLV-TR 63.99 Bis-Tris 5% sucrose 2mM MgCl2 pH 6.0 LV-MV-CMHII 61.22 PBS, 5% sucrose, 2mM MgCl2, pH 7.0 LV-MV-CMHII 65.67 PBS 5% sucrose, 2mM MgCl2, pH 7.0 LV-VSV-g 107 BISTRIS, 5% sucrose, 2mM MgCl2, pH 6.0 LV-VSV-g 104 Note: MLV-GaLV: pseudotyped murine retrovirus GaLV LV-GaLV-TR: pseudotyped lentivirus GaLV-TR LV-MV: pseudotyped lentivirus with Measles virus env (modified CMHII) LV-VSV-g: VSV-g pseudotyped lentivirus The results presented in Table 4 show that all retroviral or lentiviral vectors pseudotyped with different envelope proteins can be concentrated in the presence of sucrose and MgCl2 at pH 7.0 resulting in yields ranging from about 74% for LV-GaLV-TR to about 100% for VSVg. With respect to the use of a pH of 6.0 no difference was observed for pseudotyped VSVg vectors. For the GaLV-TR pseudotyped lentiviral vectors, these vectors were more stable at pH 7.0 during tangential filtration. The concentration / diafiltration yield was greater than 90%, while the yield was around 74% for the lentiviral vectors GaLV-TR. Anion exchange chromatography: The concentration / diafiltration step by tangential flow filtration has significantly reduced the protein and DNA load (see above) meaning that a significant portion of contaminants that could be competitors of the vectors to be purified for access to the ligands of the chromatography is decreased. In principle, depending on the subsequent use one can imagine two different ways of considering purification. They are shown in FIG. 1: a simplified method employing a single exclusion chromatography step (A in Fig. 1) and a more elaborate process using an additional anion exchange chromatography step for the preparation of lentiviral vectors for clinical use (B in Fig. 1).
[0041] The different possibilities of chromatography are developed later: Just after the TFF UF / DF step and in order to reduce the contaminants and to separate the viral particles, an anion exchange chromatography step is added. This technique makes it possible to separate the biomolecules according to their isoelectric points as a function of the pH and the concentration of salts. Thus, at a given pH value, a certain concentration of salts (often NaCl) is required in order to remove the biomolecules retained and this concentration must be chosen according to the interaction force between the biomolecules and the ligands: the more this interaction is the higher the concentration of salts (salinity) must be high. In addition, the higher the pH of the chromatography buffer is near the isoelectric point of the biomolecule species to be purified, the less salt is needed to unhook the biomolecules from the chromatographic ligands. However, it is known that retroviral and lentiviral vectors rapidly lose their infectivity as a function of salt concentration (reviewed by Segura et al., 2006). Thus, at first, the stability of the lentiviral vectors towards different concentrations of NaC1 was evaluated. 1. Impact of salinity on the stability of lentiviral GaLV-TR vectors: As indicated above, the elution of the biomolecules retained by a chromatography column is very often done with salt gradients (buffers containing NaCl) or step of increasing the salt concentration (NaCl). Therefore, in order to evaluate the effect of the NaCl concentration, incubation tests of post-TFF lentiviral vectors were carried out in different NaCl concentrations ranging from 50 mM to 1500 mM, at room temperature for 4 h. Fig. 3 represents the infectivity of the lentiviral vectors at room temperature as a function of the NaCl concentration relative to the conditions without added NaCl or the same vector preparation incubated at 4 ° C without added NaCl. This test clearly shows that a concentration of NaCl between 50mM and 1M has a moderately detrimental effect on the stability of lentiviral vectors GaLV-TR with a loss of infectivity ranging from 29.52% (50mM NaCl) to 43.86 % (1M NaCl) (percentage relative to the preparation stored at 4 ° C). On the other hand, the concentration of 1.5M NaCl leads to a 63.8% loss of infectivity when the vector preparation is stored at room temperature for 4 hours. It should be noted that storage at 20 ° C (room temperature) for 4 hours without added NaCl also leads to some loss of vector infectivity of about 23% over storage at 4 ° C. These results mean that it is essential to elute lentiviral vectors GaLV-TR chromatographic media with the lowest possible salinity, so ideally below 1M NaCl in order to maintain the maximum infectivity. In addition, it is also preferable to perform all purification (all steps) at a reduced temperature (ideally between 4 ° C and 10 ° C). 2. Evaluation of Different Anion Exchange Chromatography (AEX) Media: We used weak anion exchange chromatography media (DEAD (D)) to determine whether it was possible to limit the inactivation of the vectors. with this type of support, in particular by attempting to reduce the concentration of salt necessary to pick up said vectors from the chromatography column. In preliminary tests using a concentrated supernatant from PG13 cell cultures (MLV-GaLV) it has been possible to show that the use of a DEAE based chromatographic support (Tosoh TSK gel DEAE 5PW) leads to a yield of the infectious vector of 71% approximately higher yield than during 1 "use of a strong exchanger (Q Sepharose FF GE HealthCare) whose yield was only 16%, due to too strong interaction leading to inactivation when In this example, the salt concentration needed to unhook the retroviral vectors was 655 mM and 915 mM, respectively, and based on these results, weak anion exchangers were chosen for the rest of the study. development: several chromatography supports were evaluated: CIM D monolite (DEAE), Poros D50 (Life Technologies), Sartobind D (Sartorius) (Bandeira et al 2012), Toyopearl 650C DEAE (Merten et al., 2011). In preliminary studies, three supports were evaluated for the purification of lentiviral vectors GaLV-TR at pH 5.5 or 6.0 and 7.0. For all media tested at pH 5.5 or 6.0 and 7.0, the choice of low pH (5.5 or 6.0) was beneficial for the level of infectious vector yield: in terms of ICD D DEAE support the yield was increased from 23% (pH 7.0) to 64% (pH 6.0) when reducing the pH of buffers used for chromatography from 7.0 to 6.0 (Fig. 4). Similar results were observed for Sartobind 75D media (yield increase of 5.8% to 15.6%) and Poros D (yield increase of 32% (pH 7.0) at 80.2% (pH 6.0 and about 100% (pH 5.5), and for Toyopearl 650C gel (23% increase in yield (pH 7.0) to 89% (pH 5.5)) (Figure 4). In addition, in order to unhook the vectors of the supports, the salinity of the elution buffer could be lower during chromatography at pH 6.0 (thus, softer for the lentiviral vectors). As for the Poros D support used at pH 6.0, the elution of the vectors is at 650 mM NaCl, (see below). In terms of overall efficiency, 'modern' media (developed more recently, generating reduced shear forces (mainly due to greater porosity than other media) during chromatography and characterized by the incompressibility of the support during modification of the buffer flow rate, such as DEAE CIM monolith or Poros D) have shown higher yields than membrane (Sartobind 75D) or compressible gel (Toyopearl 650C) media. Finally, the choice fell on recent media because their efficiency of separation and recovery in vectors was higher compared to more conventional media. These two supports have therefore been more widely evaluated and their use has been optimized for the purification of lentiviral vectors. The two supports, CIM D DEAE and Poros D, have an interesting yield greater than 60%. Elution is at 650mM NaCl, Bis-Tris 5% sucrose 2mM MgCl2 pH 6.0. Increasing the pH of the elution buffer to 7.0 (PBS) results in a fall in yield of less than 7%, but the addition of 5% sucrose to PBS results in a significance increase of about 7% to 40%. %. However, it is necessary in this case to use a concentration of NaCl greater than 1M. Indeed it has been found that at pH 7.0 for the elution of the vectors (buffer without added sucrose) it will take about 1.5M NaCl in PBS, which is probably the explanation of the low yield. The negative effect of the concentration of salts on the stability of viral particles is known (Segura et al., 2005). 3. Evaluation of Different pH Values on Chromatographic Efficiency Using Poros D Support: The pH was varied within a range of 5.5 to 8.0 in the presence and absence of 5% sucrose.
[0042] The presence of 5% sucrose has a positive effect on the yield during the anion exchange chromatography step when the pH is above 5.5 (eg Poros D) (Table 5). The positive effect of the presence of sucrose on the yield is no longer observed at pH 5.5. On the other hand, the presence of sucrose is essential at pH 8.0 to recover about 58% of infectious vectors. While using pH ranging from 6.0 to 7.0, the yield is between 52 and 65%, the best yield (about 100%) is obtained at a pH of 5.5. In general, the presence of 5% sucrose leads to a reduction in the salinity necessary to initiate the elution of the lentiviral vector with a decrease in the necessary NaCl concentration of about 25 mM.
[0043] Table 5. Comparison of the yields of LV-GaLV-TR by chromatography on Poros D using buffers of different pHs (5.5 - 8.0) in the presence or absence of sucrose. With / without TU yield% sucrose pH 5.5 5% sucrose 105.98 (Bis-Tris) 0% sucrose 101.33 pH 6.0 5% sucrose 52.32 (Bis-Tris) 0% sucrose 29.29 pH 7.0 5% sucrose 65.52 (PBS) 0% sucrose pH 8.0 5% sucrose 57.76 (Bis-Tris-propane) 0% sucrose 0 4. Evaluation of an alternative protocol including anion exchange chromatography as a first step: We evaluated the yield obtained during the application of an anion exchange chromatography step immediately after the clarification step. Under these conditions, the observed yield is lower than when an ultrafiltration / diafiltration step is carried out between the clarification and the anion exchange chromatography. This latter protocol was therefore selected for further purification.
[0044] Exclusion Chromatography: Exclusion chromatography is a method of choice for the separation of biomolecules by their molecular size, thus separating particles from contaminants. Capto Core 700 filtration gel (GE HealthCare) has been used, but other media may be considered. This step allows us to replace the buffer of the previous step with the desired formulation buffer, eliminate contaminating molecules smaller than 750 kDa and avoid dilution of the sample to be loaded. This chromatography step can be directly used after tangential flow filtration (concentration / diafiltration - process A) or after an ion exchange chromatography step - process B (Fig. 1). The sample resulting from the tangential flow filtration or the sample from the fractions of the anion exchange chromatography containing the lentiviral vectors is loaded on the exclusion chromatography column. In both cases the yield of this step is 86% ± 4, depending on the fractions retained for later use. Fig. 5 shows the purification of lentiviral vectors (concentrated and diafiltered by tangential flow filtration) by gel filtration (Capto Core 700). The peak elution of the vector is at the buffer passage front and exits the column at fractions 4-9, covering about 70% of the amount of the initially loaded vectors on the column (Fig. 5a). Fig. 5B and 5C represent the analysis of each fraction by electrophoresis (Western Blot, SDSPAGE) clearly indicating the absence of the contaminating bands (FIG 5c) and the presence of the 24-25 kDa band corresponding to the capsid protein p24 of the lentiviral vector.
[0045] Yields and purities: The most important parameters concern the overall yield as well as the purity of the lentiviral vector preparation at the level of the reduction of contaminating protein and contaminating DNA load. For protocol B (including TFF, anion exchange chromatography (AEX) and exclusion chromatography (SEC)) (Fig. 1) for the purification of lentiviral vectors for clinical use, the yield is about 50% and this protocol allows 99.9% removal of contaminating proteins and 99.9% of contaminating DNA. Protocol A (comprising a TFF and exclusion chromatography (SEC)) (FIG 1) intended for the purification of lentiviral vectors for use in research is simpler because it lacks the ion exchange chromatography step. The overall yield is higher due to the reduction in the number of purification steps and reaches 60.2%, the removal of residual DNA contaminants from this simplified protocol is of the order of 96.17% and there is a reduction of contaminating proteins by 99.63%.
[0046] Practical Examples of Target Cell Transduction: Transduction of CD34 + Cells: To determine the quality of the purified vectors, CD34 + cord blood cells are transduced. The cells are thawed after 18 hours of pre-stimulation with cytokines. The transduction is for 6 hours. Then, the cells are put in a differentiation medium for 5 days. The cells were then switched to FACS FC500 (BD Biosciences) to measure the percent expression of GFP. The following results are typically obtained (Figure 6): the concentration / diafiltration purification of the lentiviral vectors (GaLV-TR) leads to an increase in the transduction efficiency of CD34 + cells (expressed as a percentage of cells expressing GFP) ranging from of 9% when using a 70% crude supernatant for use of a concentrated / diafiltered LV vector preparation. BIBLIOGRAPHIC REFERENCES - Anliker B, Abel T, Kneissl S, Hlavaty J, Caputi A, Brynza J, Schneider IC, Münch RC, Petznek H, RE Kontermann, Koehl U, Johnston IC, Keinan K, Muller UC, Hohenadl C, Monyer H , Cichutek K, Buchholz CJ (2010). Specific gene transfer to neurons, endothelial cells and hematopoietic progenitors with lentiviral vectors. Nat. Methods 7: 929-935. - Ansorge S, Henry O, Kamen A (2010) Recent progress in lentiviral vector mass production.
[0047] Biochem. Eng. J. 48: 362-377. - Bandeira V, Peixoto C, Rodrigues AF, Cruz PE, PM Alves, Coroadinha AS, Carrondo MJT (2012) Downstream processing of lentiviral vectors: releasing bottlenecks. Hmm. Gene Ther. Methods. 23: 255-263. - Charrier S, Ferrand M, Zerbato M, Precigout G, Viornery A, Bucher-Laurent S, Benkhelifa- Ziyyat S, Merten OW, Perea J, Galy A (2011) Quantification of lentiviral vector copy numbers in individual hematopoietic colony-forming cells shows vector dose-dependent effects on the frequency and level of transduction. Gene Ther. 18: 479-487. - Christodoulopoulos I, Cannon, PM (2001) Sequences in the cytoplasmic tail of the gibbon ape leukemia virus. J. Virol. 75: 4129-4138. - Greene MR, Lockey T, PK Mehta, YS Y, Eldridge PW, Gray JT, Sorrentino BP (2011) Transduction of human CD34 + repopulating cells with a self-inactivating lentiviral vector for SCID-X1 . Hmm. Gene Ther. Methods 23: 297308. - Fenard D, Ingrao D, Seye A, Buisset J, Genries S, Martin S, Kichler A, Galy A (2013) Vectofusin-1, a new viral entry enhancer, strongly promotes lentiviral transduction of human hematopoietic stem cells. Mol. Ther. Nucleic Acids 2: e90 - Frecha C, Szecsi J, Cosset FL, Verhoeyen E (2008) Strategies for targeting lentiviral vectors. Curr. Gene Ther. 8: 449-460. - Hasslacher M, Mayer C, Mitterer A (2009) Method of concentrating shear-sensitive biopolymers using hollow fiber membranes. WO patent 2010 / 025278A1 - Kutner RH, Puthli S, Marino MP, Reiser J (2009) Simplified production and concentration of HIV-1-based lentiviral vectors using HYPERFlask vessels and anion exchange membrane chromatography. BMC Biotechnol. 9: 10. - Merten, O-W (2004) State of the art of the production of retroviral vectors. J. Gene Med. 6: S105-S124. - Merten OW, Charrier S, Laroudie N, Fauchille S, Dugué C, Jenny C, Audit M, ZantaBoussif MA, Chautard H, Radrizzani M, Vallanti G, Naldini L, Noguiez-Hellin P, Galy A (2011) Large-scale manufacture and characterization of a lentiviral vector produced for clinical ex vivo gene therapy application. Hmm. Gene Ther. 22: 343-356. - Miller AD (2001) Production of retroviral vectors. Curr. Protoc. Hmm. Broom. Chapter 12: Unit 12.5. - Miller AD, Chen F.Retrovirus packaging cells based on murine 10A1 leukemia virus for the production of vectors that use multiple receptors for cell entry. J. Virol. 70: 5564-5571. - Miller, AD, Garcia, JV, Von, SN, Lynch, CH, Wilson, C., Eiden, MV (1991) Construction and properties of retroviruses packaging cells based on gibbon ape leukemia virus. J. Virol. 65: 2220-2224. - Münch RC, Mühlebach MD, Schaser T, Kneissl S, Jost C, Plückthun A, Cichutek K, Buchholz CJ (2011) DARPins: an efficient targeting domain for lentiviral vectors. Mol. Ther. 19: 686-693. - Pfeifer A, Hofmann A (2009) Lentiviral transgenesis. Methods Mol. Biol. 530: 391-405. - Rodrigues AF, Alves, PM, Coroadinha AS (2011). Production of Retroviral and Lentiviral Gene Therapy Vectors: Challenges in the Manufacturing of Lipid Enveloped Virus. Viral Gene Therapy. K. Xu, InTech. Chapter 2: 15-40. - Sakuma T, Ravin SS, Tonne JM, Thatava T, Ohmine S, Takeuchi Y, Malech HL, Ikeda Y (2010) Characterization of retroviral and lentiviral vectors pseudotyped with murine xenotropic leukemia virus-related virus envelope glycoprotein, Hum. Gene Ther. 21: 1665-1673. - Schweizer M, Merten O-W (2010) Large-scale production means for the manufacturing of lentiviral vectors. Curr. Gene Ther. 10: 474-486 - Segura MM, Kamen A, Garnier A (2006) Downstream processing of oncoretroviral and lentiviral gene therapy vectors. Biotechnol. Adv. 24: 321-337. - Segura MM, Kamen A, Trudel P, Garnier A (2005) A novel purification strategy for retrovirus gene therapy using heparin affinity chromatography. Biotechnol. Bioeng. 90: 391-404. - Stacey GN, Merten O-W (2011) Chapter 3: Hosts cells and cell banking. In: Merten O-W, Al-Rubeai M (eds.): Viral Vectors for Gene Therapy: Methods and Protocols, in the series of: Methods in Molecular Biology 737, Humana Press, New York, NY, pp 45-88.
权利要求:
Claims (18)
[0001]
REVENDICATIONS1. A method for purifying an enveloped virus, comprising an anion exchange chromatography step, wherein the buffers used in said chromatography are - pH less than 6, or - pH greater than or equal to 6 and further comprising a polyol. 10
[0002]
2. Method according to claim 1, the buffer (s) of the anion exchange chromatography being of pH less than 6 and also comprising a polyol.
[0003]
3. Method according to claim 1, the pH of the buffers being between 5.5 and 6, the pH being more particularly equal to 5.5 or 6. 15
[0004]
4. Method according to any one of the preceding claims, the anion exchange chromatography step being preceded by an ultrafiltration / diafiltration step, in particular a tangential flow filtration. 20
[0005]
5. The method of claim 4, the ultrafiltration / diafiltration step comprising the use of one or more buffers of pH between 5.5 and 7.5 optionally comprising a polyol.
[0006]
6. A method according to any one of the preceding claims for purifying a virus wrapped from the culture medium of a cell culture of cells producing said enveloped virus, the method comprising: (a) clarifying the medium; cell culture; (b) a step of ultrafiltration / diafiltration of clarified viruses; (c) anion exchange chromatography; (D) exclusion chromatography.
[0007]
7. The method of claim 6, step (a) being carried out by filtration of the culture medium on a retention filter whose retention threshold is between 0.2 and 0.45 microns.
[0008]
8. The method of claim 6 or 7, the step (b) being performed by means of a tangential flow filtration.
[0009]
The method of any of claims 6 to 8, wherein step (d) comprises using an exclusion resin having an exclusion size of between 300 and 1000 kDa.
[0010]
10. Process according to any one of claims 6 to 9, the resin used for the exclusion chromatography being multimodal, having a dual exclusion and adsorption functionality. 10
[0011]
11. Method according to any one of claims 1 to 10, the purified viruses being produced in a neutral medium or in a moderately acidic medium, in particular at a pH between pH 6 and pH 7. 15
[0012]
12. Process according to any one of claims 1 to 11, the polyol being chosen from sucrose, mannitol, sorbitol and trehalose.
[0013]
13. A process according to any one of the preceding claims wherein the polyol is present in the buffer at a concentration of between 1.5% and 15% by weight in the buffer, in particular between 2% and 5%, more particularly 5%.
[0014]
14. Process according to any one of claims 1 to 13, the polyol being present in the buffers used during a tangential flow filtration and anion exchange chromatography. 25
[0015]
15. A process according to any one of the preceding claims wherein the polyol is present in the buffers at all stages of the purification process. 30
[0016]
16. Process according to any one of the preceding claims, the buffers used during said process also comprising a magnesium salt, in particular magnesium chloride, in particular at a concentration of between 0.1 mM and 5 mM, in particular between 1 and 3 mM, more particularly at 2 mM.
[0017]
17. Method according to any one of the preceding claims, for the purification of an enveloped virus, in particular a retrovirus, in particular a lentivirus, pseudotyped with the GaLV, GaLV-TR, VSV-g or MV envelope glycoprotein.
[0018]
The process of any of claims 1 to 10 and 14 to 17, wherein the anion exchange chromatography is low anion exchange chromatography and / or column anion exchange chromatography.
类似技术:
公开号 | 公开日 | 专利标题
EP3083970B1|2019-09-04|Method for purifying enveloped viruses or viral vectors
ES2618851T3|2017-06-22|Purification of retroviral vectors
JP6480910B6|2019-04-03|An extensible production process for producing recombinant lentiviral vectors in serum-free suspension culture systems
Ansorge et al.2010|Recent progress in lentiviral vector mass production
JP2018007671A|2018-01-18|Virus-based vector composition useful for transduction of eukaryotic cells
Marin et al.2016|RD-MolPack technology for the constitutive production of self-inactivating lentiviral vectors pseudotyped with the nontoxic RD114-TR envelope
Schüle et al.2009|Restriction of HIV-1 replication in monocytes is abolished by Vpx of SIVsmmPBj
Valkama et al.2020|Development of large-scale downstream processing for lentiviral vectors
de las Mercedes Segura et al.2006|Purification and characterization of retrovirus vector particles by rate zonal ultracentrifugation
Satoh et al.2013|Gene transduction in human monocyte-derived dendritic cells using lentiviral vectors
Boudeffa et al.2019|Toward a scalable purification protocol of GaLV-TR-pseudotyped lentiviral vectors
EP3047025B1|2019-04-03|Method for producing enveloped viruses
CA2848484C|2021-03-23|Chimeric non-integrating lentiviral genomes as vaccines against hiv-1
CN103842501B|2017-12-08|viral vector purification system
US20170073702A1|2017-03-16|Virus Purification
US20210301307A1|2021-09-30|Ldlr negative packaging cell line for the production of vsv-g pseudotyped retroviral vector particles or virus particles thereof
WO2022036048A1|2022-02-17|Methods for producing clinical-grade lentiviral vector
US20180051275A1|2018-02-22|Method for purifying viral vectors
EP0273811A2|1988-07-06|Process for the preparation of a vaccine against hepatitis B
同族专利:
公开号 | 公开日
ES2753300T3|2020-04-08|
SG11201605805RA|2016-09-29|
EP3083970B1|2019-09-04|
EP3083970A1|2016-10-26|
US10465169B2|2019-11-05|
JP6553043B2|2019-07-31|
CN105980571A|2016-09-28|
FR3014901B1|2017-06-09|
US20170002332A1|2017-01-05|
DK3083970T3|2019-11-11|
JP2017503486A|2017-02-02|
WO2015092287A1|2015-06-25|
引用文献:
公开号 | 申请日 | 公开日 | 申请人 | 专利标题
WO2007123961A2|2006-04-20|2007-11-01|Wyeth|Purification processes for isolating purified vesicular stomatitis virus from cell culture|
WO2009153563A1|2008-06-18|2009-12-23|Oxford Biomedica Limited|Virus purification|EP3581646A1|2018-06-15|2019-12-18|Themis Bioscience GmbH|Integrated manufacturing and chromatographic system for virus production|FR1358909A|1962-06-13|1964-04-17|Autelca Ag|Subscriber telephone system with subscriber stations equipped with an accumulator|
JPH0253043B2|1987-11-10|1990-11-15|Tokyo Shibaura Electric Co|
WO1989006276A2|1988-01-08|1989-07-13|Dpz Deutsches Primatenzentrum Gesellschaft Mbh|Hiv-2-type retroviruses of primates, vaccines, diagnostic and pharmaceutical compositions|
CA2181066A1|1994-01-12|1995-07-20|Hitoshi Kotani|Purification of retroviral vectors|
DK1501921T4|2002-04-30|2012-10-08|Oncolytics Biotech Inc|Improved virus cleansing methods|
CN101052714B|2004-11-05|2012-01-11|威尔斯达特生物制剂公司|Stable and filterable enveloped virus formulations|
TWI469992B|2008-08-28|2015-01-21|Baxter Int|Methods of concentrating shear-sensitive biopolymers|
US10596264B2|2011-06-30|2020-03-24|Genethon|Peptides with viral infection enhancing properties and their use|
EP2782997B1|2011-11-24|2018-01-10|Genethon|Scalable lentiviral vector production system compatible with industrial pharmaceutical applications|
FR3010720B1|2013-09-16|2017-08-11|Genethon|PROCESS FOR PRODUCING ENVELOPED VIRUSES|
MX370871B|2013-12-20|2020-01-08|Dsm Nutritional Products Ag|Methods of recovering oil from microorganisms.|FR3010720B1|2013-09-16|2017-08-11|Genethon|PROCESS FOR PRODUCING ENVELOPED VIRUSES|
PT3377618T|2015-11-19|2021-01-06|Novartis Ag|Buffers for stabilization of lentiviral preparations|
EP3442688A4|2016-04-12|2019-11-20|Artemis Biosystems, Inc.|Perfusion filtration systems|
CN108524929B|2017-03-06|2019-05-07|广州瑞贝斯药业有限公司|A kind of production method of rabies vacciness|
CN107630037A|2017-10-19|2018-01-26|和元生物技术(上海)股份有限公司|A kind of purifying process for obtaining high-purity gland relevant viral vector|
AU2019286406A1|2018-06-12|2021-01-07|Kbio Holdings Limited|Virus and antigen purification and conjugation|
WO2020214676A1|2019-04-15|2020-10-22|Children's Hospital Medical Center|Viral vector manufacturing methods|
CN110499287B|2019-08-30|2021-07-23|博雅干细胞科技有限公司|Method for simply preparing placenta mesenchymal stem cell exosome|
CN110540956B|2019-08-30|2021-07-20|博雅干细胞科技有限公司|Method for simply preparing cell factor from placenta mesenchymal stem cells|
WO2021174059A1|2020-02-27|2021-09-02|Takeda Vaccines, Inc.|Method for removing host cell dna from virus preparation|
法律状态:
2015-12-08| PLFP| Fee payment|Year of fee payment: 3 |
2016-12-05| PLFP| Fee payment|Year of fee payment: 4 |
2017-12-18| PLFP| Fee payment|Year of fee payment: 5 |
2019-11-18| PLFP| Fee payment|Year of fee payment: 7 |
2020-12-10| PLFP| Fee payment|Year of fee payment: 8 |
2021-12-24| PLFP| Fee payment|Year of fee payment: 9 |
优先权:
申请号 | 申请日 | 专利标题
FR1362835A|FR3014901B1|2013-12-17|2013-12-17|PROCESS FOR PURIFYING ENHANCED VIRUSES OR VIRTORS|FR1362835A| FR3014901B1|2013-12-17|2013-12-17|PROCESS FOR PURIFYING ENHANCED VIRUSES OR VIRTORS|
PCT/FR2014/053406| WO2015092287A1|2013-12-17|2014-12-17|Method for purifying enveloped viruses or viral vectors|
SG11201605805RA| SG11201605805RA|2013-12-17|2014-12-17|Process for purifying enveloped viruses or viral vectors|
DK14828237T| DK3083970T3|2013-12-17|2014-12-17|PROCEDURE FOR CLEANING COAT-CARRYING VIRUSES OR VIRAL VECTORS|
CN201480075550.8A| CN105980571A|2013-12-17|2014-12-17|Method for purifying enveloped viruses or viral vectors|
ES14828237T| ES2753300T3|2013-12-17|2014-12-17|Purification procedure for viruses or enveloped viral vectors|
US15/105,880| US10465169B2|2013-12-17|2014-12-17|Method for purifying enveloped viruses or viral vectors|
JP2016540514A| JP6553043B2|2013-12-17|2014-12-17|Process for purifying enveloped viruses or viral vectors|
EP14828237.9A| EP3083970B1|2013-12-17|2014-12-17|Method for purifying enveloped viruses or viral vectors|
[返回顶部]