System and method for identifying viral contaminants

文档序号:1060777 发布日期:2020-10-13 浏览:15次 中文

阅读说明:本技术 用于鉴定病毒污染物的系统和方法 (System and method for identifying viral contaminants ) 是由 D·范豪特 S·戴维斯 于 2019-02-25 设计创作,主要内容包括:本公开涉及流线化的样品制备方法,即VERA(通过减少干扰物富集病毒),使总基因组材料向病毒基因组倾斜。在样品采集后8小时内即可完成这种宿主基因组干扰物的减少。使用快速文库制备方案(约15分钟)和实时纳米孔测序,可以在样品采集后不到一个工作日内鉴定出潜在的病毒污染,例如RNA病毒污染。(The present disclosure relates to a streamlined sample preparation method, namely VERA (enrichment of viruses by reduction of interferents), skewing total genomic material towards the viral genome. This reduction of host genome interferents is achieved within 8 hours after sample collection. Using a rapid library preparation protocol (about 15 minutes) and real-time nanopore sequencing, potential viral contamination, such as RNA viral contamination, can be identified in less than one working day after sample collection.)

1. A method for identifying a virus in a sample, comprising:

lysing eukaryotic cells in a sample of the cell culture;

removing cell debris from the sample;

concentrating the sample to produce a retentate;

treating the retentate with a nuclease to degrade eukaryotic nucleic acids;

extracting viral nucleic acid from the nuclease-treated retentate; and

sequencing the viral nucleic acid to identify the virus without amplifying the nucleic acid in the sample.

2. The method of claim 1, wherein the eukaryotic cell comprises a chinese hamster ovary cell.

3. The method of claim 1 or 2, wherein the eukaryotic cell secretes a protein drug product.

4. The method of claim 3, wherein the protein drug product is selected from the group consisting of an antibody or antigen-binding fragment thereof, a fusion protein, and a recombinant protein.

5. The method of claim 1, further comprising the step of treating the cell culture to remove the identified virus.

6. The method of any one of claims 1-5, wherein the cells are lysed using a freeze-thaw technique.

7. The method of any one of claims 1-6, wherein the step of removing cell debris comprises centrifuging the lysed sample to produce a supernatant, and filtering the supernatant to remove cell debris from the sample.

8. The method of any one of claims 1-7, wherein the virus is an RNA virus.

9. The method of any one of claims 1-7, wherein the virus is selected from the group consisting of Mouse parvovirus (Minutevir of mice, MVM), K virus, Mouse Encephalomyelitis virus (Mouse Enphalyelitis virus) and Mouse Adenovirus (Mouse Adenovirus), MAV1, P12, EMC, Lactate Dehydrogenase Virus (LDV), polyoma virus, Mouse Hepatitis Virus (MHV), Sendai virus, lymphocytic choriomeningitis virus (LCM), reovirus type 3, Karschhorn rat virus (Kilham rat virus), and Toolan's H-1 virus (Toolan's H-l virus).

10. A method for detecting viral nucleic acid in a cell culture sample, comprising:

lysing cells in the cell culture sample to produce cell debris;

separating the cell debris to produce a supernatant;

concentrating the supernatant to produce a retentate;

enzymatically digesting eukaryotic nucleic acids in the retentate;

extracting viral nucleic acid from the retentate;

sequencing the extracted viral nucleic acid without amplifying the extracted viral nucleic acid, wherein more viral reads are obtained from the sequencing than from the sequencing.

11. The method of claim 10, wherein the viral reads are at least 51% of the total reads obtained from the sequencing.

12. The method of claim 10, wherein the viral reads are between 50% and 99% of the total reads obtained from the sequencing.

13. The method of claim 10, wherein the viral reads are at least 80% of the total reads obtained from the sequencing.

14. The method of claim 10, wherein the viral reads are at least 85% of the total reads obtained from the sequencing.

15. The method of claim 9, wherein the viral reads are at least 90% of the total reads obtained from the sequencing.

16. The method of any one of claims 9-15, wherein the virus is an RNA virus.

17. The method of any one of claims 1-16, wherein the sequencing is completed within 8 hours of sample preparation.

18. The method of any one of claims 1-17, wherein the sequencing is real-time nanopore sequencing.

Technical Field

The present invention relates generally to methods for sequencing nucleic acids, particularly viral nucleic acid sequences, present in cell culture.

Background

Next Generation Sequencing (NGS) platforms and other High Throughput Sequencing (HTS) platforms have the potential to redefine viral safety tests in the biopharmaceutical industry. While Polymerase Chain Reaction (PCR) -based methods are sensitive, inexpensive, and rapid, NGS-based methods allow for extensive testing of all viral genomes present in a particular clinical or production sample. Heretofore, the use of this technology as a tool for systematic monitoring of potential contaminants in a production process has been hampered by the workflow required for metagenomic detection of foreign matter. Specifically, the adoption of NGS technology for conventional virus screening measures requires optimization of sample preparation strategies, streamlined and automated library preparation, and time-sensitive data analysis workflows.

Viral genomes are complex and diverse, with RNA viruses particularly complicating the preparation of adequate libraries in de novo HTS workflows. Viral contamination represents a serious threat compared to microbial and mycoplasma contamination, which can be detected relatively easily, because some viruses are difficult to detect, and also infected cell cultures lack efficient handling methods.

Therefore, new methods for detecting and identifying viral contamination in cell cultures are needed.

Disclosure of Invention

One embodiment provides a streamlined sample preparation method, namely VERA (enrichment of viruses by reduction of interferents), to skew total genomic material to the viral genome. This reduction of host genomic material can be accomplished within 8 hours after sample collection. Using a rapid library preparation protocol (about 15 minutes) and real-time nanopore sequencing, potential viral contamination can be identified in less than one working day after sample collection.

FIG. 1 is a schematic of an exemplary method for detecting and identifying viral nucleic acids in cell culture samples. A method for identifying viral nucleic acid in a cell culture sample includes lysing eukaryotic cells in a sample of the cell culture. Cell lysis may be performed using mechanical lysis techniques such as sonication or homogenization. The preferred method of lysing the cells is conventional freeze-thawing techniques. Once the cells in the cell culture are lysed, cell debris is removed from the sample. Cell debris can be removed using conventional techniques including, but not limited to, centrifugation, size filtration, or a combination thereof. Once most of the cellular debris in the sample is removed, the sample is concentrated to produce a retentate. The retentate is then treated with a nuclease to digest the eukaryotic nucleic acids. Because the viral particles protect the viral nucleic acid, nuclease treatment does not digest the viral nucleic acid. In one embodiment, with、OmniCleaveTMAnd RiboShredderTMThe retentate is treated with a combination of (a) and (b) to digest host cell DNA and RNA in the culture. After treatment with nuclease, viral nucleic acid is extracted from the retentate. Viral nucleic acids are sequenced without amplification, e.g., by real-time nanopore sequencing, and identified. In certain embodiments, more viral reads are obtained from sequencing than cell nucleic acid reads are obtained from sequencing.

In another embodiment of the disclosed method, the viral reads are at least 51% of the total reads obtained from sequencing, or from 50% to 99% of the total reads obtained from sequencing, or at least 80%, 85%, or 90% of the total reads obtained from sequencing.

Generally, the cells in cell culture are eukaryotic cells, such as chinese hamster ovary cells. In some embodiments, the cell secretes a protein drug product. The protein drug product may be an antibody or antigen-binding fragment thereof, a fusion protein, or a recombinant protein.

In certain embodiments, the virus is an RNA virus, a DNA virus, or a combination thereof. The virus may be single-stranded or double-stranded. Exemplary viruses that can be detected include, but are not limited to, Mouse parvovirus (MVM), MSV1, P12/EMC, Mouse Encephalomyelitis virus (Mouse Enphalyemylitis virus), Mouse Adenovirus (Mouse Adenovirus), Lactate Dehydrogenase Virus (LDV), polyoma virus, Mouse Hepatitis Virus (MHV), Sendai virus, lymphocytic choriomeningitis virus (LCM), reovirus type 3, Karsch rat virus (Kilham rate virus), and Toolan's H-1 virus.

Drawings

FIGS. 1A-1H are schematic diagrams of one embodiment of a method for detecting viral nucleic acid in cell culture.

FIG. 2 is a bar graph of the average number of viral reads in VERA using random primer PCR and VERA using targeted primer PCR in separate samples.

Figure 3A is a schematic showing that 95% of reads were eukaryotic (corresponding to the host) and 2% were viral when taken from the retentate. FIG. 3B is a schematic showing that 97% of reads were eukaryotic and 2% were viral when the sample was treated with nuclease.

FIG. 4 is an ethidium bromide gel showing that most of the eukaryotic DNA was removed using VERA.

FIG. 5A is a schematic view showingOmniCleaveTMAnd RiboShredderTMWhen the sample was treated with the combination of (1) 87% of the total reads were viral and 12% were eukaryotic. FIG. 5B is a schematic view showingOmniCleaveTMAnd RiboShredderTMWhen the sample was treated with the combination of (1) 92% of the total reads were viral and 5% were eukaryotic.

Detailed Description

I. Definition of

The use of the terms "a" and "an" and "the" and similar referents in the context of describing the invention (especially in the context of the following claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.

Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein.

The use of the term "about" is intended to describe values within a range of about +/-10% above or below the stated value; in other embodiments, the numerical range of values may be a range of about +/-5% above or below the stated value; in other embodiments, the numerical range of values may be a range of about +/-2% above or below the stated value; in other embodiments, the numerical range of values may be a range of about +/-1% above or below the stated value. The foregoing scope is intended to be clear from the context, and no further limitation is implied. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., "such as") provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.

The term "read" with respect to sequencing refers to a nucleic acid sequence of a set of nucleotides, which is obtained after the end of the sequencing process and is ultimately the sequence of a segment of the complete nucleic acid sequence. A "read" is a base read of a string of nucleotides derived from the original signal.

"protein" refers to a molecule comprising two or more amino acid residues joined to each other by peptide bonds. Proteins include polypeptides and peptides, and may further include modifications such as glycosylation, lipid attachment, sulfation, gamma carboxylation of glutamic acid residues, alkylation, hydroxylation, and ADP-ribosylation. Proteins may have scientific or commercial significance, including protein-based drugs, and proteins include, inter alia, enzymes, ligands, receptors, antibodies, and chimeric or fusion proteins. Proteins are produced by various types of recombinant cells using well-known cell culture methods and are typically introduced into cells by genetic engineering techniques (e.g., such as sequences encoding chimeric proteins, or codon-optimized sequences, intron-free sequences, etc.), where they can exist as episomes or integrate into the cell genome.

An "antibody" refers to an immunoglobulin molecule composed of four polypeptide chains (two heavy (H) chains and two light (L) chains) interconnected by disulfide bonds. Each heavy chain has a heavy chain variable region (HCVR or VH) and a heavy chain constant region. The heavy chain constant region contains three domains, CH1, CH2, and CH 3. Each light chain has a light chain variable region and a light chain constant region. The light chain constant region consists of one domain (CL). The VH and VL regions may be further subdivided into regions of hypervariability, termed Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, termed Framework Regions (FRs). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR 4. The term "antibody" includes glycosylated and non-glycosylated immunoglobulins of any isotype or subtype. The term "antibody" includes antibody molecules prepared, expressed, produced or isolated by recombinant means, such as antibodies isolated from host cells transfected to express the antibody. The term antibody also includes bispecific antibodies that include heterotetrameric immunoglobulins that can bind to more than one different epitope. Bispecific antibodies are generally described in U.S. patent application publication No. 2010/0331527, which is incorporated by reference herein.

"cell culture" refers to propagation or proliferation of cells in a vessel such as a flask or bioreactor, and includes, but is not limited to, fed-batch culture, continuous culture, perfusion culture, and the like.

Figure BDA0002648731490000053

Refers to the gene produced in E.coli strain W3110Genetically engineered endonuclease from Serratia autohesia (Serratiamarcescens), E.coli strain W3110 containing the proprietary pNUC 1-producing plasmid (U.S. Pat. No. 5,173,418).

OmniCleaveTMRefer to omniclearTMEndonucleases, a highly purified enzyme from recombinant strains of E.coli, degrade single-and double-stranded DNA and RNA into di-, tri-and tetra-nucleotides. The Omniclean Endonuclease (Omniclean Endonuclease) has the function of the enzyme

Figure BDA0002648731490000051

The same substrate specificity and the same product is produced,

Figure BDA0002648731490000052

is an enzyme derived from Serratia marcescens.

RiboShredderTMRefers to proprietary non-mammalian rnase blends that can completely degrade unwanted RNA during DNA and protein purification procedures. RiboShredderTMCommercially available from Epicentre corporation.

As used herein, "amplifying" refers to producing multiple copies of a DNA or RNA fragment. Amplification is usually caused by polymerase chain reaction.

As used herein, "PCR" refers to the polymerase chain reaction, a molecular biology technique for amplifying a single copy of a DNA or RNA fragment to produce thousands to millions of copies of a particular DNA or RNA sequence. PCR is commonly used to amplify the copy number of DNA or RNA fragments to be cloned, or for other analytical procedures.

The term "real-time nanopore sequencing" refers to sequencing analysis, where sequencing is performed as a result of nucleic acid molecules passing through a nanopore disrupting current flow.

Methods of detecting viruses in cell culture

A representative method of identifying and sequencing viruses present in cell culture comprises the steps of: lysing the cells in culture, removing cell debris, concentrating the sample to enrich for viral nucleic acid, enzymatically digesting nucleic acid released from the cells present in the enriched sample, extracting viral nucleic acid from the digested sample, and sequencing the viral nucleic acid to identify the virus.

A. Lysing cells

A cell culture sample is treated to disrupt or lyse the cells being cultured. Typically, the cell is a eukaryotic cell, such as a chinese hamster ovary cell. Examples of other cells and cell lines that may be used in cell culture are provided below.

Techniques for lysing cells are well known in the art. In a preferred embodiment, the cells are lysed using a freeze-thaw technique. For example, cell vials are typically frozen overnight and then thawed prior to use.

In one embodiment, the cells are lysed using mechanical means, including but not limited to a Waring Blender (Waring Blender) and Polytron. The rotating blades grind and disperse cells and tissues.

In another embodiment, the cells are subjected to Liquid Homogenization (Liquid Homogenization), for example using a Dounce Homogenizer (Dounce Homogenizer), a Potter-Elvehjem Homogenizer, or a French press. The cell or tissue suspension is sheared by forcing it through a small space.

In another embodiment, the cells are lysed by sonication using a sonicator. High frequency sound waves can shear cells.

B. Removal of cell debris

Lysing cells produces cell debris. Cell debris can be removed using conventional techniques such as centrifugation, size filtration, or a combination thereof.

1. Centrifugation

In one embodiment, cell debris is removed using low speed centrifugation. Typically, the sample is clarified by centrifuging the sample at 1,600 × g for 5 minutes. The supernatant was then removed.

2. Filtration

In one embodiment, the cell debris is removed by filtering the sample. For example, the sample can be filtered using a filter having pores with a diameter of 0.2 μm to 0.45 μm. Suitable filters are commercially available. In certain embodiments, the sample is first centrifuged and the supernatant is then filtered to remove additional cellular debris, including but not limited to organelles such as nuclei. In another embodiment, the sample is filtered using a 0.8 μm filter.

The resulting filtrate was then concentrated.

C. Concentrating the sample

Concentrating the filtrate, e.g. by using

Figure BDA0002648731490000071

100kDa concentrator and was performed according to the manufacturer's instructions. Other ways to concentrate the filtrate and increase total viral nucleic acid extraction include, but are not limited to, lyophilization. In one embodiment, the sample is concentrated by diafiltration (diafiltration).

D. Nuclease treatment

The concentrated sample is then treated with a nuclease to digest nucleic acids from cells in the cell culture. Due to the protective nature of the viral capsid, the infecting virus is immunized against the nuclease treatment of this step.

In one embodiment, with

Figure BDA0002648731490000081

OmniCleaveTM、RiboShredderTMAnd combinations thereof to treat the concentrated sample. RNAse Cocktail (RNAse Cocktail Mix) (RNAse Cocktail Mix) can also be usedRNaseCocktailTMIs a mixture of two highly purified ribonuclease RNase A (500U/mL) and RNase T1(20,000U/mL).

In one embodiment, the two virus filtrate volumes are distributed independently on Amicon 15 microconcentrators with 100kDa passable filters. If necessary, the tubes can be equilibrated using a phosphate buffered saline solution and centrifuged at maximum speed on an Allegra 6 centrifuge for 15 minutes. The retentate volume was checked. If necessary, the tube was centrifuged again at maximum speed for 15 minutes. The volume of retentate in each microconcentrator should be about 200. mu.L. The retentate was carefully removed from each microconcentrator and pooled in a new tube. A total retentate of 50. mu.L was removed from each sample and labeled sample DP and sample DR.

1 XDNase buffer was added to the two separate pooled retentates to increase the volume to 4 mL. Concentrate in Amicon 4 as done in step 4.4, but reduce the volume to 50 μ L to 200 μ L.

To each volume of buffer exchanged retentate, 2 volumes of omniclean, 2 volumes of Benzonase, 2 volumes of RNase Mix (RNase Cocktail Enzyme Mix) were added and sufficient 10 x dnase buffer was added to make the final 1 x dnase buffer. The tubes were incubated at 1000 rpm for 2 hours at 37 ℃ in a thermostatic mixer.

Other nucleases that can be used include, but are not limited to, DNase I, DNase II, nuclease S1, RNase A, RNA, enzyme I, RNase T1, RNase T2, RNase H, exonuclease I, exonuclease II, exonuclease III, exonuclease IV, exonuclease V, lambda exonuclease, endonuclease I, endonuclease II, endonuclease III, endonuclease IV, endonuclease V, endonuclease VI, endonuclease VII, endonuclease VIII and site specific endonucleases including, but not limited to, AatII, Acc, AclI, AatII, Acc65I, AccI, AclI, Afei, AflII, AgeI, ApaI, ApaLI, ApoI, AscI, AseI, AsiHrII, AvrII, BamHI, BclI, BglII, Bmel580, Bsam I, Bsa389, BsiI, BsitBII, EsIII, EascIII, HagIII, EaszsIII, EsIII, ClasIII, ClasHisII, ClasIII, ClasII, EsIII, ClasIII, BspI, EsIII, BspI, EsIII, BspI, EsIII, BspI, E, KasI, KpnI, MfeI, MluI, MscI, MspAlI, MfeI, MluI, MscI, NaeI, NarI, NcoI, NdeI, NgoMIV, NheI, NotI, NruI, NsiI, NspI, PacI, PciI, PmeI, PmlI, PsiI, PspOMI, PstI, PvuI, PvuII, SacI, SacII, SalI, SbfI, ScaI, SfcI, SfoI, SgrAI, SmaI, SnaBI, SpeI, SphI, SspI, StuI, SwaI, XbaI, XhoI, and XmaI.

In one embodiment, the sample is treated with one or more nucleases at 37 ℃ for digestion for 2 hours on a shaker (i.e., a homomixer). 2 Xnuclease the retentate was sufficient (2 XOmnicarave, 2 XBenzonase, 2 XRNase mix). In addition, 10 XDNase buffer was added to a final concentration of 1X. Thus, different sample volumes can be handled by easily scaling up or down the ratio of enzyme to retentate.

E. Extraction of viral nucleic acids

After digestion with the nuclease mixture, viral nucleic acid is extracted from the sample. In one embodiment, 0.5M EDTA is added to the tube to a final concentration of 3mM EDTA prior to extraction. PureLink from ThermoFisher, Seimer, Inc. can be usedTMViral RNA/DNA minikit (PureLink)TMViral nucleic acid was extracted using Viral RNA/DNA MiniKit).

To prepare the Wash Buffer (Wash Buffer), 60mL of 96-100% ethanol was added to 15mL of the Wash Buffer (WII) included in the kit. The wash buffer label was checked to indicate that ethanol was added and was at room temperature.

Carrier RNA was prepared using 5.6 μ g of Carrier RNA per sample (for <500 μ L samples). Less Carrier RNA may be used per sample, but the amount of Carrier RNA required for each sample type and downstream application should be verified.

Preparation of Carrier RNA (5.6. mu.g/sample):

1. mu.L of RNase-free Water (RNase-free Water) (included in the kit) was added to 310. mu.g of lyophilized Carrier RNA provided in a tube together with the kit to obtain 1. mu.g/. mu.L of Carrier RNA stock.

2. Mix well and aliquote the solution into small portions. Aliquots were stored at 20 ℃.

Repeated freeze thawing is avoided.

3. The volume of lysis buffer (lysbuffer)/Carrier RNA mixture required to simultaneously treat the required number of samples was calculated using the following formula:

n × 0.21mL (lysis buffer volume/reaction) ═ a mL

A mL×28μL/mL=BμL

Wherein

Number of samples N

Calculated volume of lysis buffer (L22)

B ═ calculated volume of 1 μ g/μl Carrier RNA stock to be added to lysis buffer (L22)

4. Required amount of 1. mu.g/. mu.L Carrier RNA stock was thawed.

5. In a sterile tube, the volume of Carrier RNA stock (B, calculated as described above) was added to the volume of lysis buffer (a, calculated as described above). Mix gently by pipetting up and down. Vortex is avoided as it creates foam.

6. Storage at 4 ℃ until use. Buffer was used within 1 hour.

Preparation of lysate

Described below for 200L starting material lysate preparation scheme. If you wish to treat > 200. mu.L (< 500. mu.L) of sample volume, please enlarge the reagent volume accordingly.

1. Add 25. mu.L proteinase K to a sterile microcentrifuge tube.

2. To this microcentrifuge tube was added 200. mu.L of cell-free sample.

Note that: if you are handling <200 μ L of sample, please adjust the final volume of the sample to 200 μ L using PBS (phosphate buffered saline) or 0.9% NaCl.

3. Add 200. mu.L lysis buffer (containing 5.6. mu.g Carrier RNA). The tube was capped and vortex mixed for 15 seconds.

4. Incubate at 56 ℃ for 15 minutes.

5. Add 250 μ L96-100% ethanol to the tube, cap and vortex mix for 15 seconds.

6. The lysate was incubated for 5 minutes at room temperature.

Purification of

1. The above lysates were added to a Viral Spin Column (Viral Spin Column) in a collection tube.

2. The column was centrifuged at 6800 Xg for 1 min. The collection tube was discarded. The spin column was placed in a new wash tube (WashTube).

3. The column was washed with 500. mu.L of washing buffer (WII) containing ethanol. Centrifuge at 6800 Xg for 1 min. The flow-through stream is discarded.

4. The washing step 3 was repeated using 500. mu.L of washing buffer (WII) each time.

5. The collection tube was discarded and the spin column placed in another clean wash tube.

6. The spin column was centrifuged at maximum speed for 1 minute to remove any residual wash buffer (WII).

7. The column was placed in a clean 1.7mL Recovery Tube (Recovery Tube).

8. Elute with 10-50 μ L sterile rnase-free water (E3) attached to the kit (water added to the center of the cartridge).

9. Incubate at room temperature for 1 minute. The column was centrifuged at maximum speed for 1 minute to elute the nucleic acid. The recovery tube contains purified viral nucleic acid. Discard the spin column.

10. The purified viral RNA/DNA is stored at-80 ℃ or the RNA/DNA is used for the desired downstream applications.

F. Sequencing

Once viral nucleic acid is extracted, library preparation can begin for sequencing. Importantly, viral nucleic acids are not amplified prior to sequencing. In one embodiment, library preparation is performed using SQK-LSK308(1D2) or SQK-RAD004(Rapid) ONT library preparation kits, according to the manufacturer's instructions. SQK-RAD004 was a 15 minute rapid library preparation kit, while SQK-LSK308 was a slower (about 3 hours) more robust library preparation.

In a preferred embodiment, a third generation sequencer (MinION) from Oxford nanopore technologies, Inc. (Oxford Nanopore technologies) is usedTM) Sequencing the viral nucleic acid. MinIONTMThe portable small-sized sequencer can generate sequencing data in real time, so that the time for identifying pollutants received from a sample is greatly reduced. Native sequencing of foreign substance nucleic acids also eliminates the need for time-consuming (targeted or de novo) amplification steps. Using the WIMP software of the ONT to analyze the data, the platform can compare the data of the reads to the virus, bacteria and fungi database from RefSeq and update it periodically. Corresponding "eukaryotic" or "fineBacterial "alignment" represents a false positive most likely derived from partial alignment of CHO (host) reads with fungal (eukaryotic) databases or contamination from reagents.

In a preferred embodiment, the viral nucleic acid is sequenced by RT-PCR. As used herein, "RT-PCR" refers to the reverse transcription polymerase chain reaction, RT-PCR being a variant of traditional PCR. RT-PCR is a molecular biology technique for quantitatively detecting gene expression by generating complementary dna (cdna) transcripts from RNA. During RT-PCR, RNA molecules are converted into their cDNA sequence by reverse transcriptase. The primers used for cDNA synthesis may be non-sequence specific primers (such as a mixture of random hexamer primers or oligo dT primers) or sequence specific primers. Random hexamers are mixtures of all possible combinations of hexanucleotide sequences that can be randomly attached to an entire RNA pool. Oligo dT primers are complementary to the poly-A tail of the mRNA molecule and can be used to synthesize cDNA from mRNA molecules only. Traditionally, cDNA synthesized from mRNA was amplified using standard PCR procedures.

Viruses that can be detected and sequenced using the disclosed methods include, but are not limited to, hantavirus, lymphocytic choriomeningitis virus, rat rotavirus, reovirus type 3, sendai virus, mouse podofungual disease virus, K virus, krestin rat virus, P12, EMC, MAV1, lactate dehydrogenase virus, mouse parvovirus, tirel's virus, mouse hepatitis virus, mouse rotavirus, polyoma virus, rat coronavirus retrovirus, sialorrhagitis virus, thoracoadenovirus, HI virus, murine leukemia virus, and bovine viral diarrhea virus.

Human viral pathogens that infect cell cultures include, but are not limited to, adenovirus, cytomegalovirus, enterovirus, herpes simplex virus, influenza virus, parainfluenza virus, rhinovirus, respiratory syncytial virus, and varicella zoster virus.

Proteins of interest

The cells in the cell cultures disclosed herein typically express or secrete a protein of interest. Any protein of interest suitable for expression in prokaryotic or eukaryotic cells can be used in the provided engineered host cell systems. For example, proteins of interest include, but are not limited to, antibodies or antigen-binding fragments thereof, chimeric antibodies or antigen-binding fragments thereof, ScFv or fragments thereof, Fc fusion proteins or fragments thereof, growth factors or fragments thereof, cytokines or fragments thereof, or extracellular domains of cell surface receptors or fragments thereof. The protein of interest may be a simple polypeptide consisting of a single subunit, or a complex multi-subunit protein comprising two or more subunits. The protein of interest may be a biopharmaceutical product, a food additive or a preservative, or a protein product subject to purification and quality standards.

In some embodiments, the protein product (protein of interest) is an antibody, human antibody, humanized antibody, chimeric antibody, monoclonal antibody, multispecific antibody, bispecific antibody, antigen-binding antibody fragment, single chain antibody, diabody, triabody or tetrabody, Fab fragment or F (ab')2 fragment, IgD antibody, IgE antibody, IgM antibody, IgG1 antibody, IgG2 antibody, IgG3 antibody or IgG4 antibody. In one embodiment, the antibody is an IgG1 antibody. In one embodiment, the antibody is an IgG2 antibody. In one embodiment, the antibody is an IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG4 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1 antibody. In one embodiment, the antibody is a chimeric IgG2/IgG1/IgG4 antibody.

In some embodiments, the antibody is selected from the group consisting of: anti-programmed cell death 1 antibodies (e.g., anti-PD 1 antibodies as described in U.S. patent application publication No. US2015/0203579a 1), anti-programmed cell death ligand 1 (e.g., anti-PD-L1 antibodies as described in U.S. patent application publication No. US2015/0203580a 1), anti-dii 4 antibodies, anti-angiopoietin 2 antibodies (e.g., anti-ANG 2 antibodies as described in U.S. patent application publication No. 9,402,898), anti-angiopoietin-like 3 antibodies (e.g., anti-AngPtl 3 antibodies as described in U.S. patent No. 9,018,356), anti-platelet derived growth factor receptor antibodies (e.g., anti-PDGFR antibodies as described in U.S. patent No. 9,265,827), anti-Erb 3 antibodies, anti-prolactin receptor antibodies (e.g., anti-PRLR antibodies as described in U.S. patent No. 9,302,015), anti-complement 5 antibodies (e.g., anti-TNF C5 antibodies as described in U.S. patent application publication No. US2015/0313194a 1), anti-epidermal growth factor receptor antibodies (e.g., anti-EGFR antibodies as described in U.S. patent No. 9,132,192 or anti-EGFRvIII antibodies as described in U.S. patent application publication No. US2015/0259423a 1), anti-proprotein convertase subtilisin 9 antibodies (e.g., anti-PCSK 9 antibodies as described in U.S. patent No. 8,062,640 or U.S. patent application publication No. US2014/0044730a 1), anti-growth and differentiation factor 8 antibodies (e.g., anti-GDF 8 antibodies, also known as anti-myostatin antibodies, as described in U.S. patent No. 8,871,209 or 9,260,515), anti-glucagon receptors (e.g., anti-GCGR antibodies as described in U.S. patent application publication No. US2015/0337045a1 or US2016/0075778a 1), anti-VEGF antibodies, anti-IL 1R antibodies, interleukin 4 receptor antibodies (e.g., anti-IL 4 antibodies as described in U.S. patent application publication No. US 2015/588 a 6866 or U.S. patent No. 8,735,095 or 8,945,559)'.S. 201429, Anti-interleukin 6 receptor antibodies (e.g., anti-IL 6R antibodies as described in U.S. patent nos. 7,582,298, 8,043,617, or 9,173,880), anti-IL R antibodies, anti-interleukin 33 (e.g., anti-IL R antibodies as described in U.S. patent application publication nos. US 2014/R a R or US 2014/R a R), anti-respiratory syncytial virus antibodies (e.g., anti-RSV antibodies as described in U.S. patent application publication nos. US 2014/R a R), anti-differentiation cluster 3 (e.g., anti-CD R antibodies as described in U.S. patent application publication nos. US 2014/R a R and US R a R), anti-CD 3620 (e.g., anti-CD R antibodies as described in U.S. patent application publication nos. US 2014/R a R and US R a R), anti-CD R antibodies as described in U.S. patent application publication nos. US 2014/R a 36, Anti-cluster of differentiation 48 (e.g., anti-CD 48 antibody as described in U.S. patent No. 9,228,014), anti-Fel d1 antibody (e.g., as described in U.S. patent No. 9,079,948), anti-middle east respiratory syndrome virus (e.g., anti-MERS antibody as described in U.S. patent application publication No. US2015/0337029a 1), anti-ebola virus antibody (e.g., as described in U.S. patent application publication No. US 2016/0215040), anti-sakavirus antibody, anti-lymphocyte activation gene 3 antibody (e.g., anti-LAG 3 antibody or anti-CD 223 antibody), anti-nerve growth factor antibody (e.g., anti-NGF antibody as described in U.S. patent application publication No. US2016/0017029 and U.S. patent nos. 8,309,088 and 9,353,176), and anti-activin a antibody. In some embodiments, the bispecific antibody is selected from the group consisting of: anti-CD 3 x anti-CD 20 bispecific antibodies (as described in U.S. patent application publication nos. US2014/0088295a1 and US20150266966a 1), anti-CD 3 x anti-mucin 16 bispecific antibodies (e.g., anti-CD 3 x anti-Muc 16 bispecific antibodies), and anti-CD 3 x anti-prostate specific membrane antigen bispecific antibodies (e.g., anti-CD 3 x anti-PSMA bispecific antibodies). In some embodiments, the protein of interest is selected from the group consisting of: abciximab (abciximab), adalimumab (adalimumab), adalimumab-atto (adalimumab-atto), ado-trastuzumab (ado-trastuzumab), alemtuzumab (alemtuzumab), aleurozumab (aleuromab), aleurizumab (atezolizumab), avizumab (avelumab), basiliximab (basiliximab), belimumab (belimumab), benralizumab (benralizumab), bevacizumab (bevacizumab), belotoxmumab (bezlotoxumumab), bornatuzumab (blinatumomab), benitumomab (brentuximab), brentuzumab (brentuzumab), natamycin (brentuximab), properitumumab), propertizumab (propertizumab), propertizumab (propertilizumab), netuzumab (brentuximab), brentuximab (bretuzumab), najikulimumab (prazelizumab), propertib (propertib), propertizumab (pegucizumab), and (pegucizumab (pegucidoxab), and (pegucizumab (netucidoxumab), and (e), and (netucizumab (praecoxib), e (praecoxib), e (e), e (praecoxib), e (e), and (praecoxib), e (e), e (e), e (, Rituximab (elotuzumab), emmenizumab (emilizumab-kxwh), ertatin-alilizumab conjugate (emtansineairumab), elvucinoguman mab (eintacumab), elolimumab (einuiuzumab), fatinumab (einimumab), golimumab (gusimumab), cekuzumab (gusukukumab), ibritumomab (ibritumomab tiuxetan), idazukizumab (idarubizumab), infliximab (infliximab-abda), infliximab-dyb (infliximab-ydab), ipilimumab (infliximab), ibritumomab (infliximab-abda), infliximab-dyb (infliximab-ydab), ipilimumab (ipilimumab), ibritumumab (iximab), martensib (polemeuzumab), emmuzumab (inozumab), emmenizumab (inomab), inolizumab (inozumab), inolizumab (inomycin), infliximab (inomab), inomycin (inomycin), infliximab (orizumab), ibritumab (orimab (orizumab), orimazomab (oribivakub), ibritumab (inomab (oxepirubib (inomab), ibritumab (inomab), ibritumumab (inobruxib), ibrinomab (inobruxib (inomab), ibrinomab (inobruxib), eukamex-niuzumab), ibrinobruxib (inobruxib), eukamex-mex-niuzumab), ibritumumab (inomaben-niuz, Pembrolizumab (pembrolizumab), pertuzumab (pertuzumab), ramucirumab (ramucirumab), ranibizumab (ranibizumab), resibazumab (raxibacumab), rayleigh-based mab (resilizumab), linumab (rinucumab), rituximab (rituximab), sarilumab (sarilumab), secukinumab (secukinumab), cetuximab (siltuximab), tacitumab (tocilizumab), trastuzumab (trastuzumab), tevacizumab (trovuzumab), umumab (ustekkinumab), and vedolizumab (vedolizumab).

In some embodiments, the protein of interest is a recombinant protein (e.g., an Fc fusion protein) that contains an Fc portion and another domain. In some embodiments, the Fc fusion protein is a receptor Fc fusion protein comprising one or more extracellular domains of a receptor coupled to an Fc moiety. In some embodiments, the Fc portion comprises the hinge region of IgG, followed by CH2 and CH3 domains. In some embodiments, the receptor Fc fusion protein contains two or more distinct receptor chains that can bind to a single ligand or multiple ligands. For example, Fc fusion proteins are TRAP proteins such as, for example, IL-1 TRAPs (e.g., rilonacept, which contains an IL-1RAcP ligand binding region fused to the extracellular region of IL-1R1 fused to the Fc of hIgG 1; see U.S. Pat. No. 6,927,004, which is incorporated herein by reference in its entirety) or YEGF TRAPs (e.g., affibercept or ziv-affibergept, which contains Ig domain 2 of VEGF receptor Flk1 fused to Ig domain 3 of VEGF receptor Flk1 fused to the Fc of hIgG 1; see U.S. Pat. Nos. 7,087,411 and 7,279,159). In other embodiments, the Fc fusion protein is an ScFv-Fc fusion protein that contains one or more of one or more antigen binding domains of an antibody (such as a variable heavy chain fragment and a variable light chain fragment) coupled to an Fc portion.

Cell culture

The cell culture described herein may be a "fed-batch cell culture" or a "fed-batch culture," which refers to a batch culture in which cells and medium are initially supplied to a culture vessel and additional culture nutrients are slowly fed to the culture in discrete increments during the culture, with or without periodic cell and/or product harvesting prior to terminating the culture. Fed-batch culture includes "semi-continuous fed-batch culture" in which the entire culture (possibly including cells and medium) is periodically removed and replaced with fresh medium. Fed-batch culture is distinguished from simple "batch culture" in which all the components for cell culture (including animal cells and all culture nutrients) are supplied to the culture vessel at the beginning of the culture process. Fed-batch culture may differ from "perfusion culture" in the following respects: in standard fed-batch processes the supernatant is not removed from the culture vessel, whereas in perfusion culture the cells are confined in the culture by e.g. filtration and the medium is continuously or intermittently introduced into and removed from the culture vessel. However, it is envisaged that samples are removed for testing purposes during fed-batch cell culture. The fed-batch process is continued until it is determined that the maximum working volume and/or protein yield is reached, followed by harvesting of the protein.

The cell culture may be a "continuous cell culture," which is a technique for continuously culturing cells, typically in a particular growth phase. For example, if a continuous supply of cells is required, or if a particular protein of interest is to be produced, the cell culture may need to be maintained in a particular growth phase. Therefore, conditions must be continuously monitored and adjusted accordingly to maintain the cells in this particular growth phase.

Cells are cultured in cell culture medium. The terms "cell culture medium" and "culture medium" refer to a nutrient solution used to culture mammalian cells that typically provides the necessary nutrients to enhance the growth of the cells, such as carbohydrate energy sources, essential amino acids (e.g., phenylalanine, valine, threonine, tryptophan, methionine, leucine, isoleucine, lysine, and histidine) and non-essential amino acids (e.g., alanine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, proline, serine, and tyrosine), trace elements, energy sources, lipids, vitamins, and the like. The cell culture medium may contain an extract, such as serum or peptone (hydrolysate), which may provide a feedstock to support cell growth. The culture medium may contain an extract derived from yeast or soybean, instead of an extract derived from animals. Chemically defined media refers to cell culture media in which all chemical components are known (i.e., have a known chemical structure). Chemically defined media are completely free of animal derived components such as serum derived or animal derived peptones. In one embodiment, the medium is a chemically defined medium.

The solution may also contain components that enhance growth and/or survival above a minimum rate/ratio, including hormones and growth factors. The solution can be formulated to a pH and salt concentration that is optimal for the survival and proliferation of the particular cells being cultured.

By "cell line" is meant one or more cells derived from a particular lineage by serial passage or subculture of the cells. The term "cell" is used interchangeably with "cell population".

The term "cell" includes any cell suitable for expression of a recombinant nucleic acid sequence. Cells include prokaryotic and eukaryotic cells, such as bacterial cells, mammalian cells, human cells, non-human animal cells, avian cells, insect cells, yeast cells, or cell fusions, such as, for example, hybridomas or quadruple hybridomas. In certain embodiments, the cell is a human, monkey, ape, hamster, rat, or mouse cell. In other embodiments, the cell is selected from the following cells: chinese Hamster Ovary (CHO) (e.g., CHO K1, DXB-11CHO, Veggie-CHO), COS (e.g., COS-7), retinal cells, Vero, CV1, renal cells (e.g., HEK293, 293EBNA, MSR 293, MDCK, HaK, BHK21), HeLa, HepG2, WI38. MRC 5, Colo25, HB 8065, HL-60, lymphocytes such as Jurkat (T lymphocytes) or Daudi (B lymphocytes), A431 (epidermal cells), U937, 3T3, L cells, C127 cells, SP2/0, NS-0, MMT cells, stem cells, tumor cells and cell lines derived from the above cells. In some embodiments, the cell comprises one or more viral genes, such as a retinal cell (e.g., PER) that expresses a viral gene.

Figure BDA0002648731490000191

A cell). In some embodiments, the cell is a CHO cell. In other embodiments, the cell is a CHOK1 cell.

24页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:通过芳香族化合物增强核酸聚合

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!