AU653656B2 - Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications - Google Patents

Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications Download PDF

Info

Publication number
AU653656B2
AU653656B2 AU29756/92A AU2975692A AU653656B2 AU 653656 B2 AU653656 B2 AU 653656B2 AU 29756/92 A AU29756/92 A AU 29756/92A AU 2975692 A AU2975692 A AU 2975692A AU 653656 B2 AU653656 B2 AU 653656B2
Authority
AU
Australia
Prior art keywords
hiv
expression
cells
expression vector
antisense
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU29756/92A
Other versions
AU2975692A (en
Inventor
Horst-Peter Dr. Antonicek
Jorg Dr. Baumgarten
Lothar Dr. Biesert
Thomas-Peter Dr. Hausner
Axel Dr. Kretschmer
Antonius Dr. Loebberding
Burkhard Dr. Mielke
Helga Prof. Dr. Rubsamen-Waigmann
Wolfgang Dr. Springer
Udo Dr. Stropp
Mark-Michael Dr. Struck
Harry Dr. Suhartono
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bayer AG
Original Assignee
Bayer AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to DE4126484A priority Critical patent/DE4126484A1/en
Priority to EP92118414A priority patent/EP0594881A1/en
Priority to CS923337A priority patent/CZ284778B6/en
Priority to NZ24523092A priority patent/NZ245230A/en
Priority to CA002083812A priority patent/CA2083812A1/en
Application filed by Bayer AG filed Critical Bayer AG
Priority to AU29756/92A priority patent/AU653656B2/en
Priority to JP4343223A priority patent/JPH06153958A/en
Publication of AU2975692A publication Critical patent/AU2975692A/en
Application granted granted Critical
Publication of AU653656B2 publication Critical patent/AU653656B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16211Human Immunodeficiency Virus, HIV concerning HIV gagpol
    • C12N2740/16222New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/16011Human Immunodeficiency Virus, HIV
    • C12N2740/16311Human Immunodeficiency Virus, HIV concerning HIV regulatory proteins
    • C12N2740/16322New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Virology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Description

The present invention relates to the targeted blocking of the genetic information of the viral messenger ribonucleic acid (mRNA) for virus replication of HIV) in transfected, haematopoietic cells with particularly suitable expression systems, which express a complementary "antisense RNA".
In particular the invention relates to expression vectors coding for antisense RNA sequences, which, in contrast to the state of the art, produce complete inhibition of t 10 HIV-1 replication and clear inhibition of HIV-2 replication in human cells which have been altered using gene technology. The genetic units described here of the expression vectors are particularly suited for the somatic gene therapy of HIV/AIDS diseases, because they protect haematopoietic cells from HIV-induced cytopathic effects. Immune insufficiency in HIV-infected humans can thereby be avoided.
Virus multiplication in an infected cell is regulated by the genes of the virus genome. In this process, regulatory gene products and structural proteins for the construction of infectious virus particles for virus multiplication are formed by these virus genes by means of transcription and translation. The essential intermediate for this process i. messenger ribonucleic acid (mRNA), which mediates the information for the formation of the regulatory nd structural proteins.
Le A 28 518 1 .e Le A 28 518 1 o o* Blocking of the messenger ribonucleic acid in a highly specific manner, which rests entirely on the uniqueness of the molecular structure of the viral mRNA, namely the virus-specific nucleotide-base sequence of this molecule, can be achieved with a ribonucleic acid of complementary base sequence: by sequence-specific nucleotide-base pairing, as has been described in an analogous manner for deoxyribonucleic acid, the previously single-stranded mRNA is complexed in a double-stranded RNA, and the genetic information of the nucleotide-base sequence H becomes in part unavailable for protein biosynthesis. The second step of gene expression, i.e. translation, is thereby inhibited. The complementary ribonucleic acid is termed antisense RNA.
The mechanism of the inhibition of gene expression by interactions of the antisense RNA with the target mRNA is complex and to a large extent unclarified. One explanation is the inhibition of RNA splicing and RNA processing in the cell nucleus; detailed investigations on this point are however not known. Evidence of adenosine inosine exchange in double-stranded RNA has been published, and inhibition of translation suggested as a consequence of this exchange. The inhibition of RNA transport through the perinuclear membrane to the site of 25 protein biosynthesis and the masking of the ribosome/RNA interaction by double-stranded RNA may provide further contributions to the inhibition of gene expression by :antisense RNA.
Le A 28 18 Le A 28 518 2- The inhibition of metabolic processes by antisense RNA has been described in naturally occurring biological systems, such as bacteria and some eukaryotic, multicellular organisms Takayama and M. Inouye, Critical Reviews in Biochemistry and Molecular Biology 25, 155 (1990)], as well as in transgenic organisms produced artificially by gene transfer Mol et al. FEBS Letters 268, 427 (1990)].
There have also been reports on attempts in plants Cuozzo et al., Biotechnology 6, 549 (1988)) and animal cells Riden and E. Gilboa, J. Virol. 63, 677 9 (1989), EP 252 940] to produce virus-resistant organisms following the introduction of a gene expressing antisense RNA. There are however no convincing, scientifically proven connections between antisense RNA expression and virus resistance, as the authors themselves state in these publications. In particular, in the case of microinjected plasmids expressing antisense RNA with transient expression the anti-viral activity is weak or leads to anti-viral effects which cannot be conclusively evaluated Rittner et al., Nucleic Acids Research 19, 1421 (1991)]. There has also been a report on the use of retrovirus vectors expressing antisense RNA To et al. in: Gene Regulation: Biology of Antisense RNA and DNA ed. R.P. Erickson and J.G. Izant, Raven Press Ltd., New York 1992] for producing virus-resistant cells. These retroviral vectors carry the risk of triggering malignant tumour growth, because they contain oncogene-like gene Le A 28 518 3 S Co o o eoo sequences, and there is multifarious evidence for retrovirus-dependent oncogenesis.
In contrast to the published works on antisense RNA for controlling virus replication, expression constructs are described in the present invention which are distinguished by the particular nature of their promoter constructs in combination with the specially chosen regions of the viral DNA in the antisense orientation, and which thereby provide anti-viral effects of a quality and quantity which are surprising and which surpass by far the scope which has been known hitherto. The I combination of expression vector components: DNA sequence, promoter, insert/antisense DNA and 3'processing signal sequences led, for the first time with regard to retroviral infections, to quite pronounced and clearly demonstrable virus resistance in transfected cells expressing antisense RNA. By contrast, the state of the art with other vector systems for endogenous antisense RNA expression and retrovirus resistance results in weak anti-viral effects, which only appear transiently T. Riden and E. Gilboa, J. Virol, 63, 677 (1989), EP 252 940, G. Sczakiel et al., Biochem.
Biophys. Res. Com. 169, 643 (1990) 0. I. Mirosknichenko et al., Gene 84, 83 (1989), Shimada et al., Antiviral Chemistry and Chemotherapy 2, 133 (1991); G. Sczakiel et al. J. Virol. 66, 5576 (1992) or is affected by the abovementioned risks (retroviral gene transfer) or is impractical because microinjections are necessary.
Le A 28 518 o The invention relates to expression vector constructs for the expression of antisense RNA which contain hybrid promoter gene sequences and possess a strong constitutive promoter activity or a promoter activity which can be induced in the case of pathogen attack.
These are, for example, those expression vector constructs which contain subgenomic gene sequences of a pathogenic vizus in the orientation (antisense orientation of .ie strand) under the control of the promoter for the expression of antisense RNA.
Very suitable in the scope of the present invention are those which contain proviral, subgenomic DNA in the antisense orientation from retroviruses such as HIV-1 and HIV-2 or from oncoviruses such as HTLV-I, HTLV-II or from oncogenes such as abl, erb A, erb B, fms, fos, myb, myc, ras, sis and src.
Preferred are expression vector constructs which contain HIV-1 proviral DNA in the antisense orientation of the nucleotides 7514-474 or 5786-474 or 2096-474 or 3825-681 or 5786-4158 or 5746-4648 or 2369-1635 or 3226-3003 or 2099-678 or 3829-2099 or 4647-4157 or 4157-3820.
O
Expression vector constructs may also be employed which contain subgenic proviral gene sequences or oncogene sequences in the 3'15' orientation for the expression of 25 shorter antisense-RNA transcripts.
Le A 28 518 0 Such expression vector constructs which are to be mentioned here are those which contain proviral gene sequences from HIV-1 of the nucleotides 605-455 or 670-440 or 825-535 or 6070-5800 or 5640-5020 or 5600-+5040 or 8690-8300 or 9160-8800.
Preferred are expression vector constructs which contain two, three or more subgenomic gene sequences from one virus or from a plurality of viruses.
The invention also relates to transfected cells which contain the abovementioned expression vector constructs.
The use of the said expression vector constructs for the preparation of medicaments is also within the scope of the invention as are medicaments containing one or more of the expression vector constructs.
Using the herein-described expression vector constructs, an anti-viral effect for HIV replication was demonstrated in relevant human host cells that provides complete protection against HIV-1 replication in human cells for over 60 days and additionally produces significant inhibition of HIV-2 replication.
O In the present invention, the construction and composition are described of expression vector constructs o.
which express messenger ribonucleic acid transcripts in transfected cells, which transcripts are complementary to mRNA sequences of unwanted viral genes, using here as Le A 28 518 6 6 examples the genes GAG, POL, VIF, REV, TAT and VPU [W.A.
Haseltine et al., Scientific American USA, 1988, 34 to 42], of the human immunodeficiency virus, HIV-1. In addition, the antisense-RNA sequence described is sufficiently homologous towards HIV-2 isolate HIV-2 D194, in: Human Retroviruses and AIDS, eds. Myers et al.
Los Alamos National Lab., 1992, and EMBL accession No.
X52223) in the region of the genes GAG, POL and VIF to achieve antiviral effects against HIV-2 isolates as well.
The expression vector constructs are distinguished by the particular nature of the arrangement of selected promoter sequences and hybrid promoter sequences CMV-IE/ i metallothionein I) with subgenomic HIV-1 DNA fragments in the antisense orientation which result in the transcription of several antisense-RNA transcripts by RNA splicing in a constitutive or an inducible manner.
Antisense-RNA transcripts formed in this way showed in a surprising manner a drastic or complete inhibition of HIV-1 replication in human cells (HUT 78, U 937 for example). A clear inhibition of HIV-2 replication was also observed. HIV-2 demonstrates sufficient RNA-sequence homology in the expression vector constructs described here to ensure that inhibition of virus replication by antisense RNA is brought about. The invention thus comprises a method for inhibiting the replication of retroviruses, using as examples HIV-1 and HIV-2, which is suitable for protecting haematopoietic cells, and thereby important cells for the human, cell-mediated immune response T-cells, monocytes), from cell-damaging 30 and cell-disintegrating virus multiplication by a
*A
*Le A 28 518 7 *e ft** particular mode of expression of antivirally active antisense RNA.
In order to make cells of the immune system resistant to the cell-damaging effect of retrovirus multiplication, as it is known from HIV-1 and HIV-2 infection, by means of the expression of anti-viral antisense RNA, the experimental procedure was as follows.
Promoters: Expression vectors were constructed which contain pro- 10 moters which should express constitutively and strongly in haematopoietic cells, such as, for example, cytomegalovirus IE promoter (CMV-IE). Furthermore, promoters were also constructed, by the fusion of nucleic acid sequences, which should additionally induce amplified expression in the case of virus attack. In particular, a component of these hybrid promoters is the CMV-IE metallothionein promoter fragment and the HIV-1 LTR nucleic acid fragment of the nucleotides 289 to 474 or 289 to 532 (nucleotide numbering: UWGCG GENEMBL DATA BANK File: HIVHXB2CG. VIRAL of the 25th Sept. 1987), which contributes to amplified, optionally transactivatable, supplementary promoter activity.
The antisense nucleic acid fragments Additionally the vector constructs contain subgenomic, 25 proviral DNA fragments in the antisense orientation, that Le A 28 518 8is, the (+)-strand of the DNA is transcribed in the direction, under the control of the described promoters.
For safety reasons a complete, proviral genomic DNA fragment of a retrovirus is not used. However, longer, subgenomic, proviral DNA fragments rather than short, subgenic, proviral DNA fragments are used in the main, because RNA splicing is more likely to occur in longer RNA transcripts and, in this way, more antisense RNA transcripts are possibly to be expected from the proviral DNA fragments according to the invention, and were indeed found in the examples mentioned. (The type and number of the completely processed antisense mRNA transcripts that are to be expected for the HIV sequences that were used in the antisense orientation is not predictable, because there are no sufficiently accurate consensus sequences for the splicing of mRNA that would permit a conclusive prediction.) With an increasing number of antisense RNA transcripts, with their inherent properties, such as, for example, export from the nucleus or stability in the cytoplasm, there is also an increasing probability of producing an antisense RNA transcript with potent antiviral activity.
The invention relates additionally to expression vector constructs which contain a fused gene fragment for the 25 transcription of antisense RNA under the control of a strong promoter. The fused gene fragment is composed of subgenomic DNA fragments from two different or several Le A 28 518 9
V
different pathogenic viruses, e.g. from HIV-1 and HIV-2.
Using such fused gene fragments, which encode an RNA transcript containing several antisense-RNA sequence regions one after the other which are complementary to mRNA-sequence regions from different viruses, expression vectors can be constructed with a wider anti-viral effect.
The validity of this principle is confirmed by Examples 1, 4, 5, 10 and 12. For example, the expression vector constructs KTX 11, pSXK1 and pSXK2 contain antisense-DNA fragments from HIV-1 LAV/BRU, which themselves contain shorter segments which are also complementary to mRNA from HIV-2 D194 nucleotides 620-660 or 837-899 or 4868-5044).
In this way the antisense RNA from KTX11, pSXK1 and pSXK2 is also anti-viral towards HIV-2.
The antisense RNA-encoding sequence of these vectors can be additionally fused with fragments of proviral DNA in the antisense orientation from other viruses HIV-2 isolates, other HIV-1 isolates), so that, as a result of the segementally optimised complementarity of the anti- Ssense RNA to the sequence of the other viruses, improved or complete inhibition of different viruses can be e0 achieved at the same time with one expression vector construct.
Termination of the antisense-RNA transcripts is achieved Le A 28 518 10 S «o
S
C C*
S
Le A 28 518 10 S S with the polyadenylation signal from SV 40 or from the bovine growth hormone), in order to generate a highly stable mRNA. Additionally, the vectors contain the complete operon for expression of a suitable selection marker. In the examples presented, an antibiotic resistance TNR 5 neo (neomycin, geneticin, G 418, kanamycin) is used in order to be able to select cells which after transfection contain the genetic construct for antisense- RNA expression.
This selection marker has proved to be of value for human haematopoietic cells. Other selection markers, such as, for example, puromycin or hygromycin or methotrexate in O combination with transfection of the gene for dihydrofolate reductase, could also be used.
Particularly suitable as antisense nucleotide sequences are those from the POL/VIF gene region of HIV between nucleotides 2100-5800. In particular, antisense nucleotide sequences of the nucleotides 5786-4158 have been shown to have exceptional anti-viral activity. Other fragments from this gene region can also be used, e.g.
5746-4648 or 4647-4157 or 5880-.3880 or 5850-4158 or 5150-3200.
In order to demonstrate the anti-viral effect of the antisense RNA against, for example, HIV-1, human cell 25 lines which can be infected with HIV-1 were transfected with the abovementioned expression vector constructs. The aim was to demonstrate in a series of cell types of human Aees **Le A 28 518 11 9 *0 o ooo• haematopoietic cells that endogenous antisense RNA expression leads to inhibition of virus replication in cells which in particular participate in the cellular immune response. This destruction of cells of the immune system in the terminal phase of the AIDS disease has a causal association with the increased frequency of opportunistic infections, which then often have a fatal outcome. For this reason, the aim of the work with haematopoietic cells, and in this context initially with permanent cell lines such as, for example, HUT 78, MOLT 4, U 937, Jurkat, CEM-CM3 (all American Type Culture Collection ATCC, Bethesda, Maryland, USA), was, following vector transfection, cloning of the cell lines and genetic characterisation, to determine the "virus resistance" of these transfected cell lines in infection experiments with appropriate HIV-1 and HIV-2 isolates.
The intention was that demonstration of the expression of antivirally active antisense RNA in these cells should indicate the usefulness of the expression constructs in blood-forming cells and in the end also their use for the transformation of cells of the blood-forming bone marrow tissue for therapeutic purposes. The incorporation of genes into peripheral blood lymphocytes and bone marrow cells for therapeutic purposes has already been described elsewhere W. Culver et al., Transplant Proc. 23, 170 177 (1991)] and should in principle be applicable tc the herein-mentioned antisense RNA expression constructs.
L1 e* 6* 4 Examples Example 1 Expression vector construct KTX 11 (In the following, all nucleotide numbering relates to the human immunodeficiency virus type 1 (HXB2) sequence, UWGCG, GENEMBL DATA BANK, File: HIVHXB2CG.VIRAL of the Sept. 1987, unless otherwise indicated.) For the endogenous expression of anti-virally active antisense RNA following transfection of HIV-1 infectable cells, the following expression vector was constructed: The murine metallothionein I promoter [1600 base pairs EcoRI Bgl II fragment Glanville et al., Nature 292, 267 269 (1982))] was ligated with the Sal I cleavage site (nucleotide 5786) from the 3' region of the proviral HIV-1 DNA sequence following ligation of a Bgl II-Xho I adapter, resulting in modification of the Bgl II, Xho I and Sal I cleavage sites.
The Bgl II cleavage site (nucleotide 474) from the J region of the proviral HIV-1 DNA was ligated with a Bam HI cleavage site, to which the sequence of the SV polyadenylation signal is connected (nucleotides 2564 to 4713 with deleted sequences from the UWGCG GenEMBL File SV 40XX). This same polyadenylation sequence from SV is used in the direction for the termination of the A 2 e* *o *o*oo* o Le A 28 518 13 :d [selection marker neomycin (phosphotransacetylase), nucleotide 1 1286 TRN5NEO BACTERIA, UW GCG GenEMBL]. The neomycin gene expression is initiated by the SV 40 early promoter (nucleotides 5171-371, UWGCG GenEMBL SV 40XX). Otherwise the vector also contains pBR 322 DNA (nucleotides 2067-4363, UWGCG GenEMBL File PBR 322) with bacterial origin and ampicillin resistance gene, in order to be able to use it as a shuttle vector for E. coli and animal cells. A gene map of this vector KTX 11 is presented in Figure 1.
Example 2 9 Expression vector construct KTX 12 The HIV-1 antisense-RNA expression vector KTX 12 is constructed in the same way as vector KTX 11 (Example 1).
However, it contains an inducible hybrid promoter and the HIV-1 template DNA strand extends in the orientation behind this hybrid promoter from nucleotides 75144474.
To obtain the hybrid promoter, the HIV-1 LTR promoter fragment nucleotide 289 to 532 was fused onto the Q metallothionein promoter nucleotide 368 (UW GCG File MUSMETI.RO). A restriction map of this vector KTX 12 is presented in Figure 2.
Le A 28518 14 S. S
°S.
Example 3 Expression vector construct KTX The HIV-1 antisense RNA expression vector KTX 15 is constructed in a similar way to the vector KTX 12 (Example However, the DNA fragment of the HIV-1 template strand (in the orientation) is substantially shortened, extending from nucleotide 2096 to nucleotide 474 and thereby comprising the GAG sequence, the leader sequence, the U5 sequence and a part of the R sequence. A further important property of vector KTX is the deletion of the TAR sequence with the deletion of nucleotides 474-532 in the HIV-LTR hybrid promoter. As a consequence, it should still be possible to activate the hybrid promoter in the case of HIV infection, but the TAR-encoded termination of transcription Y. Kao et al., Nature 330, 489 (1987)] without the presence of the HIV-1 TAT protein should be prevented. This hybrid promoter is thereby distinguished by a particularly high constitutive transcription under the control of an HIV-1 LTR promoter sequence. A restriction map of the vector KTX 15 is presented in Figure 3.
Example 4 Expression vector construct pSXK 1 The HIV-1 antisense-RNA expression vector pSXK 1 has been 25 prepared in order to show expression of antisense RNA e o Le A 28 518 15 under the control of another strong, constitutively expressing promoter, the example here being the cytomegalovirus promoter. The HIV-1 gene fragment nucleotide 681-3825 was inserted in the orientation into the Hind III restriction site at nucleotide 891 (vector pRc/CMV, Catalogue No. V 750-20 from Invitrogen, San Diego, CA 92121, USA). The polyadenylation signal comes from the bovine growth hormone (BGH) gene.
Otherwise the vector is constructed as a shuttle vector in the same way as vector KTX 11 (Example 1) with the neomycin selection marker and a pBR322 DNA fragment for O replication and selection in E. coli. A restriction map of vector pSXK 1 is presented in Figure 4.
Example Expression vector construct pSXK 2 The HIV-1 antisense-RNA expression vector pSXK 2 differs from vector pSXK 1 (Example 4) in that it possesses fused promoter sequences comprising nucleotides 209-865 of the CMV promoter (see Example fused with nucleotides 150- 369 of the metallothionein promoter (UWGCG GenEMBL File ^V MUSMETI.RO) and thus also represents a hybrid promoter.
In addition, the proviral DNA sequence of the nucleotides 4158-5786 was inserted in the orientation as the or* template for the antisense-RNA expression under the Goo 25 control of the CMV/metallothionein hybrid promoter. The restriction map of vector pSXK 2 is reproduced in Figure SLe A 28 518 *e
S
Example 6 Expression vector constructs pHTT 1, 4, 6, 9, 10, 12 and The HIV-1 antisense-RNA expression vectors pHTT 1, 4, 6, 9, 10, 12 and 15 were prepared in a similar manner to vector pSXK 1 (Example 4, Fig. All the HIV-1 gene fragments inserted into the pHTT vectors were cloned in the orientation into vector pRc/CMV (from Invitrogen, San Diego, CA 92121, Catalogue No. V 750-20, 1991), under the control of the constitutively expressing cytomegalovirus promoter.
The pHTT vectors are specified below: pHTT 1: The HIV-1 gene fragment (nucleotide 4648- 5746) was incorporated into the Not I cleavage site (nucleotide 966) of the pRc/CMV vector.
pHTT 4: The HIV-1 gene fragment (nucleotide 1635- 2639) was incorporated into the Xba I cleavage site (nucleotide 985) of the pRc/CMV vector.
pHTT 6: The HIV-1 gene fragment (nucleotide 3003- 3227) was incorporated into the Xba I cleavage site as described under Le A 28 518 17 Le A 28 518 17
S.
0o* 4 o e- ooe e e pHTT 9: The HIV-1 gene fragment (nucleotide 678-2099) was incorporated into the Xba I cleavage site as described under pHTT 10: The HIV-1 gene fragment (nucleotide 2099- 3829) was incorporated into the Xba I cleavage site as described under pHTT 12: The HIV-1 gene fragment (nucleotide 4157- 4647) was incorporated into the Xba I cleavage site as described under pHTT 15: The HIV-1 gene fragment (nucleotide 3830- 4157) was incorporated into the Xba I cleavage site as described under A general diagram of the HIV-1 gene fragments which are contained in the antisense orientation in the vectors of Examples 1 to 6 is reproduced in Figure 6.
Example 7 Transfection and cloning of human lymphocytes with antisense-RNA expression vectors The non-adherently growing, monocyte-like cell line U 937 20 and the non-adherently growing T-lymphocyte cell line HUT 78 were chosen as examples of transfected human haematopoietic cell lines, which exhibit virus resistance as a consequence of endogenously expressed antisense RNA Le A 28 518 18 against HIV-1. Both cell lines were obtained from the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, MD 20852 1776, USA.
The cell lines were cultured in the medium recommended by the ATCC. The vector transfection was effected by various methods of which electroporation proved to be the most successful: 1. Calcium phosphate method: A variation of the method of Wigler et al., 1977, Cell 11, 223.
The transfection is carried out with Ca precipitates of the DNA. Varying quantities from 0.05 to 20 pg of DNA were dissolved in 0.25 ml of 250 mM CaC1 2 and slowly pipetted into 0.25 ml of 2 x HEBS buffer.
Transfection was achieved by addition of the whole precipitate mixture to 10 ml of cell culture. hours later the medium was removed by suction. The cells were then shocked with DMSO.
For a 10 ml culture, 3 ml of a 25% DMSO solution in sterile PBS were added. After 4 min. the liquid was removed by suction and the cells were washed with ml of PBS. Subsequently the cells were taken up O in 10 ml of DMEM medium.
5 2. DEAE-dextran 25 On the day before the transfection, the cells were subcultured to a density of 3-4 x 105 per ml of o Le A 28 518 -19culture medium. 2-4 h before the transfection, the cells were centrifuged and taken up in fresh medium.
to 20 pg of DNA were mixed with 1070 pl of PBS; to this were added 120 pl of a 10 mg/ml DEAE-dextran solution (transfection solution). Before the transfection, 40 ml of the cells were centrifuged, taken up in 10 ml of PBS and centrifuged once again.
The cell pellet was suspended in 1.2 ml of PBS-DEAEdextran solution and incubated for 30 min. Subsequently the cells were shocked by addition of 0.8 ml O of cold DMSO solution (25% in TBS). After 3 min.
ml of TBS were added and the mixture was centrifuged. The cells were washed in RPMI growth medium and taken up in 40 ml of the same medium.
3. Fusion (Sanari-Goldin method) Logarithmically growing cells (1 x 10 7 cells/ml) were centrifuged and washed once with RPMI medium. 2 ml of a protoplast suspension of E. coli (2 x 3 cells/ml in 10% sucrose and 10 mM MgCl 2 were added to the cell pellet. The protoplasts with the transfecting plasmid were prepared by the method of Sanari-Goldin et al. After 8 min. of incubation, centrifugation was carried out and PEG solution (polyethylene glycol 400; 45% solution in RPMI) was 25 then carefully added within 2 min. After 1 min. of incubation, 10 ml of RPMI medium without FCS were added within 7 min. The cells were centrifuged, .i.
*0 Le A 28 518 a ee washed in RPMI medium and taken up in 10 ml.
4. Electroporation Electroporation (EP) was carried out with the gene pulser apparatus and capacitance extender from Bio Rad. Logarithmically grown cells were adjusted to a cell density of 1 x 10 7 cells/ml, washed 1 x in PBS and resuspended in RPMI medium without FCS with mM dextrose and 0.1 mM dithiothreitol.
0.4 ml of the cell suspension were placed in an EP cuvette and 10 20 pg of DNA were added. The DNA was dissolved in the proportion of 1 pg of DNA per 1 pl of TE.
The electroporation conditions were varied as follows: Capacitance: 260-960 pF; voltage 150-300 V; electrode distance 0.2/0.4 cm; resistance 100-m ohm.
Before and after the electroporation, the cells were incubated for 10 min. at room temperature, and then 20 subcultured in 10 ml of growth medium 10% FCS.
e e.
Selection of transfectants: The selection of transfectants was in each case carried Le A 28 518 21 0 0 Le A 28 518 21 0 out on the basis of resistance to G 418.
1. Selection in viscous media.
The transfected cells were first of all preincubated in G 418-containing growth medium until no further growth was observable. After that they were sown in viscous medium. Either agar (Gibco) or methylcellulose was used for solidification of the medium. The pre-selected cells were sown onto a tissue culture plate (10 cm), and to this were added 10 ml of selection medium (RPMI 1 mg/ml G 418) and 2 ml of a 2% liquefied agar solution. After the agar had cooled, the plates were incubated in the incubator at 37 0 C. In a variation of this experiment, the agar was replaced with methylcellulose.
2. Selection with subsequent cloning in a Transwell cell culture chamber (Transwell TM, from Costar) 48 hours after the transfection, the cells were sown on a 96-well microtitre plate at a density of 1 x 10 5 cells per 0.2 ml well. G 418 was added at a concentration of 0.7 mg/ml for U 937 and 1.0 mg/ml for HUT 78. As soon as the medium was exhausted, 1/2 the volume of each well was exchanged for fresh medium; in the case of U 937 only a further a.
0.5 mg/ml of G 418 was then added.
S 25 Incubation was continued until resistant cells were Le A 28 518 22 Le A 28 518 22beginning to grow in the individual wells. These mixed clones were expanded to a larger volume and subsequently cloned. For this, a special growth chamber from Costar (Transwell TM, Catalogue No.
3425, 205 Broadway, Cambridge, MA 02139, USA) was employed.
The cells were sown at low cell density (50 to 200 cells per 6 ml) in soft agar with G 418 (see Selection in viscous medium) and 6 ml were added to a central chamber (upper compartment). The chamber is separated by a membrane from a very thickly growing fluid feeder culture of U 937 or HUT 78 (200,000 cells/ml) (lower compart- W ment). The fluid medium is partially exchanged after exhaustion, in order to permit further cell growth. As soon as G 418-resistant colonies appeared in the soft agar, they were isolated with a Pasteur pipette and expanded.
In this way, some ten independent, transfected cell lines were cloned, for U 937 and HUT 78, with each of the vectors KTX 11, KTX 12, KTX 15, pSXK 1 and pSXK 2, and further processed for the demonstration of antisense-RNA expression and HIV-1 resistance. In addition, HUT-78 cell lines were generated with the vectors pHTT1, pHTT4, pHTT9 a and pHTT10 and examined in HIV-infection tests.
LeA2851823 ft .:.iLe A 28 518 23 4 *oo Example 8 Transformation of bone marrow cells with the expression vector KTX By way of example, the basic procedure for demonstrating transformation of primary bone marrow cells in the mouse model was as follows: Primary bone marrow cells were isolated from the femurs of six-to ten-week old C57 mice. The cells were taken up A in McCoy medium (from Flow) and mixed with interleukin-3 and GM-CSF (granulocyte-monocyte colony stimulating factor; from Genzyme), in each case at a concentration of Ag/ml, and agar (Bacto agar, from Difco; final concentration and plated out at a concentration of 1 x 105 cells per ml in the presence or absence of the toxic antibiotic G418 (neomycin) in Petri dishes. A transfection was carried out directly after the removal of the bone marrow cells before plating out in agar.
Subsequently the cells were cultured for several days at 37 0 C in the incubator and at 7.5% CO,. After various time intervals, the number of transformed colonies (at least cells) or clusters (10 to 50 cells) were in each case 4 counted under the microscope. Under the given experimental conditions, most of the colonies were mixed granulocyte/monocyte or granulocyte colonies; erythroid 25 bursts were occasionally observed, as were megakaryocytes on one occasion. The identification of the cell types in the colonies was carried out by May-Grinewald staining.
Le A 28 518 24 P P The transfection conditions were optimised. Successful transfections of primary bone marrow cells were obtained under the following conditions: 800 pl of bone marrow cell suspension (5 x 106 cells in electroporation medium as described in Example 5 pg of KTX15, electroporation at 300 V, 100 0, 250 pFD, 6 ms.
After the electroporation (gene pulser apparatus and capacitance extender from Bio Rad), the cells were selected in the abovementioned culture medium in the presence of 500 pg/ml G-418 (neomycin). Resistance to G- 418 indicated successful transfection of propagatable primary bone marrow cells- Example 9 Demonstration of the integration of intact expression vector constructs and demonstration of antisense-RNA expression Following disruption of the transfected U 937 and HUT 78 call lines, the DNA and RNA from these cell lines was isolated by standard methods using caesium chloride density-gradient centrifugation G. Davis et al., Basic Methods in Molecular Biology, Elsevier, New York (1986)1. Demonstration of the integration of intact *S vector constructs for antisense-RNA expression was carried out by means of the polymerase chain reaction Le A 28 518
S*
S" Le A 28 518 25 *o*o* [PCR Technology Principles and Application for DNA Amplification, Ed. Henry A. Erlich, M. Stockton Press, New York (1989)] using oligonucleotide primers which amplified overlapping DNA fragments of promoter sequence, insert sequence and termination sequence. Subsequently the amplified DNA was identified by means of Southern blot analysis (Davis et al., see above) using DNA insert gene probes. Examples of analytical results are given in Figures 7, 8 and 9. For example, the flanking primers 5'-CAA ACC CTT TGC GCC CG-3' or 5'-ACT CGT CCA ACG ACT AT-3' from the promoter sequence and 5'-TTT TTT CAC TGC ATT CTA CTT-3' from the polyadenylation sequence were used for the vectors KTX 11, KTX 12 and KTX 15. In the case of the expression vectors with the pRc-CMV vector, the flanking primers 5'-CTT TCC AAA ATG TCG TAA CAA CTCC-3' and 5'-ATT TAG GTG ACA CTA TAG AAT-3' were, for example, used for the promoter sequence and for the termination sequence, respectively. In the insert (HIV sequence) primer sequences were chosen which yielded PCR products of about 500 base pairs to 2,100 base pairs depending on the choice of primer pairs.
Demonstration of antisense-RNA expression was carried out by Northern blot analyses (Davis et al., see above) of RNA from the cloned, transfected cell lines. An example of an analytical result for the demonstration of HIV-1 antisense RNA in a transfected U 937 cell line is given in Figure
*A
go •e •loo fo Example Demonstration of the virus resistance of transfected, cloned haematopoietic cell lines Cloned, antisense RNA-expressing cell lines, which were obtained and characterised according to Examples 7 and 9, were examined for the expression of virus resistance in comparative infection studies with HIV-1 Ratner et al., Nature 313, 277 (1985)] or HIV-2 D 194 KUhnel et al., Nucleic Acids Research (1990) 18, 6142) together with the starting cell lines HUT 78 and U 937 as controls.
1. Infection of HUT 78 and U 938 cells with HIV-1 (controls) Procedure In each case 10 ml of cell suspension were treated for 60 min. with different dilutions of HUT 78/HIV-1 culture supernatant (about 104 SFU/ml in PBL cultures, RT activity of the frozen stock solution 650,000 cpm/ml). The cells were subsequently centrifuged off and washed three times in order to remove unbound virus particles and virus proteins. Finally the cells were in each case resuspended in 10 ml of culture medium and cultured in the incubator.
25 Every 3 to 4 days, 200 pl of the cell-free culture SS S •Le A 28 518 27 V, ft supernatant were tested for the presence of HIV antigens. An antigen-capture assay from Organon Technika was used.
To maintain the cells, 5 ml of cell suspension were in each case removed and replaced with 5 ml of culture medium.
2. Infection of antisense RNA-expressinq HUT 78 cells and U 937 cells with HIV-1 Procedure In each case 10 ml of cell suspension (about 4 x cells/ml) were treated for 60 min. with diluted culture supernatant from HUT 78/HIV-1 (about 104 SFU/ml in PBL cultures, RT activity of the frozen stock solution 650,000 cpm/ml); the final dilution of the stock virus solution was 1:1,000 for the HUT 78 cells and 1:50 for the U 937 cells. The cells were subsequently centrifuged off and carefully washed four times in order to remove unbound virus particles and virus proteins. Finally the cells were resuspended in each case in 10 ml of culture medium (RPMI 1640 16% FCS) and cultured for 60 days in the incubator.
Every 3 to 4 days, 160 1p of the cell-free culture supernatant were tested for the presence of HIV 25 antigens. An antigen capture assay from Organon Le A 28 518 28 r0 0 Technika was used with the accompanying microtitre plate photometer.
To maintain the cells, 5 ml of cell suspension were in each case removed every 3 to 4 days and replaced with 5 ml of fresh culture medium.
As representative results for other HIV antisense RNAexpressing cell lines, two series of experiments with three HUT 78 transfectants (HUT K14-KTX11, SSHUTK16/ 2-SXK1, SSHUTK1/1-SXK2) (see Fig. 11) and with three U 937 transfectants (ssUK 20/4-KTXlla, ssUK 3/1-SXK2 and N ssUK 20/15-KTBllb) (Fig. 12) may be mentioned which show no indications of HIV multiplication after infection during an incubation period of up to 60 days in these antisense RNA-expressing cell lines harbouring the expression vectors pSXK1 and pSXK2, whether reverse transcriptase activity (RT activity) or an HIV antigen is measured in these cell cultures. By contrast, HIV multiplication is clearly measurable in the control cell line. Additionally, a delayed increase in virus replication can be observed in transfectants with the more weakly expressing expression vector KTX11. The result is presented in Figures 11 and 12.
In order to check for HIV-1 infection having occurred in the cell lines SSU K3/1-SXK2, SSHUT K16/2-SXK1 and SSHUT 25 K1/1-SXK2, in which no HIV-1 antigen or RT activity could be detected after 60 days of incubation, positive identification was demonstrated, for example, by PCR a a.
Le A 28 518 29 8 a *o a detection of HIV-1 TAT gene sequences and envelope gene sequences in the cells of SSHUT K16/2-SXK1 and SSHUT K1/.-SXK2 which had been incubated for 60 days (Fig. 13).
For this purpose the PCR primers 5'-ATG GAG CCA GTA GAT CCT-3' and 5'-TCT ACC ATG TCAT-3' were used to generate a 690 base pair-long PCR fragment. The expression vectors pSXK1 and pSXK2 do not contain these sequences and cannot therefore yield any false-positive PCR product.
Example 11 Control experiments Transfection of the vectors from Examples 1 to 6 without the inserts of proviral DNA from HIV-1 in the antisense orientation did not, in the cell lines U 937 and HUT 78, lead to cell lines in which virus replication was inhibited. Control experiments with the vectors from Examples 1 to 6, in which, however, the proviral HIV-1 DNA fragment is inserted in the sense orientation, were not carried out, because the corresponding sense transcripts can bind reyulatory proteins of HIV replication, such as, for example, TAT and REV, and thereby inhibit virus replication competitively J. Graham et al., PNAS 87, 5817 (1990)] and thus appear unsuitable as control experiments.
ee
S.
e *0 Le A 28 518 30 S Example 12 Infection of antisense RNA-expressing HUT 78 and U937 cell lines with HIV-2 Infection experiments were carried out with the isolate HIV-2 D194 Kiihnel et al., Nucleic Acids Res. 18 (1990) 6142). The virus strain suspension with an RT activity of 2.14 x 105 cpm/ml and an antigen concentration (1:1,000 dilution) of 0.249 OD 450 o (160 pl sample) was stored at -80 0 C. 50 1p of this virus strain suspension were employed for the infection of 2 x 106 HUT 78 cells in 4 ml of culture medium, which was incubated for 3 hours.
In the case of U937 cells, 2 x 106 cells were incubated for the infection with 100 pl of virus strain suspension in 4 ml of culture medium for 24 hours. Subsequently, unbound virus particles were removed from the cell culture by washing.
At intervals of 3 to 4 days the cell cultures were maintained and cell culture supernatant was removed for quantitative HIV antigen determination with the HIV antigen-capture assay from Organon Technika in association with the accompanying microtitre plate photometer.
Q The course of HIV-2 D194 replication in the HUT 78 control cell lines and U937 control cell lines, which do *o not contain antisense-RNA expression vectors, is compared 25 in Figures 14 and 15 with that in the transfected cell lines, which demonstrate delayed HIV-2 multiplication as a result of antisense RNA-expression.
8 a Le A 28 518 31- Results With the expression vectors KTX 11, KTX 12, KTX pSXK 1, pSXK 2, pHTT 1, pHTT 4, pHTT 6, pHTT 9, pHTT pHTT 12 and pHTT 15 (Examples 1 to and following gene transfection and cloning of transfected cell lines (Example antisense RNA against HIV-1 was formed endogenously in human blood cell lines (Example 9) and its antiviral effect was demonstrated (Examples 10, 12).
It is surprising that a clearly expressed inhibition of the replication of the retrovirus HIV-1 was demonstrated H in the transfected cell lines. The virus resistance of the antisense RNA-expressing cells could also be shown in a similar manner with an HIV-1 virus which was specially adapted to the transfected cell lines (Fig. 11, 12). A marked antiviral property of the antisense RNA was also observed against HIV-2 (Fig. 14, These results show, by way of example, that suitable promoters for the expression of antisense RNA molecules, and naturally also the nature of the antisense-RNA sequences, lead to the development of resistance against retrovirus attack in genetically altered cells. It was shown in Example 9 that, because of previously unknown RNA splicing of HIV-1 RNA transcripts of the (+)-strand of the proviral HIV-1 DNA in the orientation, 25 several mRNA molecules arise which lead to the anti-viral effect.
Le A 28 518 32 C
C.
o* Le A 28518 32- 0l**« iio** *~ea By means of the experiments which were described in Examples 1 to 12, it was shown that, using suitable expression vector constructs with antisense RNA-encoding DNA fragments from a retrovirus, antiretroviral properties can be produced in cells and these properties protect against retrovirus attack.
From Figures 11 to 13, it is evident that the cell lines SSHUT K16/2-SXK1 and SSHUT K1/1-SXK2 are, 60 days after HIV infection, positive for the detection of proviral HIV DNA by means of PCR analysis, but negative as regards the detection of HIV antigen. This result leads to the conclusion that, following HIV-1 infection of the cells, proviral DNA is indeed still formed in the transfected HUT 78 cell lines in the first step of the HIV replication cycle, but that the transcription/translation of proviral DNA is prevented by the antisense RNA and no HIV protein for virus replication is formed in detectable amounts.
In the DNA of the cell line SSU K3/1-SXK2, neither the formation of HIV proteins or proviral HIV DNA could be detected in any of the test samples, using the antigen test and PCR analysis, respectively, at 60 days after O infection with HIV-1. This result may be interpreted as indicating that the antisense RNA formed in the cell line 25 SSU K3/1-SXK2 not only suppresses the second step in the S. HIV replication cycle (the transcription and translation of proviral DNA), but also even inhibits the first step in the replication cycle: Le A 28 518 33 .i*
S
Le A 28 518 33 o
*J~
The formation of proviral DNA from the genomic (+)-strand RNA by the process of reverse transcription can evidently be prevented by antisense RNA. This effect of antisense RNA has been speculatively assumed, but could not be demonstrated previously To and P.E. Neiman in Gene Regulation: Biology of Antisense RNA and DNA eds.
R.P. Erickson and J.G. Izant, Raven Press Ltd., New York 1992), because correspondingly powerful expression vector constructs have not hitherto been discovered.
.i t 9..
9@*9 *99* 09 9 3.
9* t a. S 49939, 0 0 Le A 28 518 34 Legends Figure 1 Expression vector KTX 11 for the expression of HIV-1 antisense RNA. The metallothionein promoter (MTI promoter) drives the transcription of antisense RNA from a proviral HIV-1 DNA fragment of the nucleotides 5786-474 (numbering: UWGCG GENEMBL DATA BANK File HIV HXB2CG.
VIRAL]. The antisense-RNA transcripts are terminated by the SV 40 polyadenylation sequence. The neoycin resistance gene TRN5neo is transcribed by the SV 40 early Q promoter. The shuttle vector also contains the pBR322 origin and the ampicillin gene (pBR 322-amp). Further details are described in Example 1.
Figure 2 Expression vector KTX 12 for the expression of HIV-1 antisense RNA under the control of an HIV-1 LTR metallothionein hybrid promoter. Details of the construction of the vector are described in Example 2.
Figure 3 Expression vector KTX 15 for the expression of HIV-1 antisense RNA under the control of an HIV-1 LTR metallothionein hybrid promoter, in which the HIV-1 TAR sequence is deleted in order to achieve maximum expression without the presence of the transactivator protein TAT. Details Le A 28 318 aa o t* S of the construction of the vector are described in Example 3.
Figure 4 Expression vector pSXK 1 for the expression of HIV-1 antisense RNA under the control of the cytomegalovirus promoter (CMV). Details of the construction of the vector are described in Example 4.
Figure Expression vector pSXK 2 for the expression of HIV-1 antisense RNA under the control of the cytomegalovirus promoter (CMV). The proviral HIV-1 DNA fragment from the central genomic region of HIV does not, in this example, contain sequences of the 5' region as in Examples 1 to 4, but instead a sequence from the POL/VIF region. Details of the construction of the vector are given in Example Figure 6 Basic diagram for the preparation of HIV-1 gene fragments, which are contained in the antisense orientation (arrow direction) in the antisense-RNA expression 20 vectors with the designation given in column 1. The promoter of the respective expression vector is indicated in column 2, and the nucleotide numbering of the HIV sequence according to UWGCG GENEMBL DATA BANK File: HIVHXB2CG.VIRAL is indicated in column 3.
**Le A 28 518 36 e* *o••e e• Figure 7 Southern blot analysis, following PCR amplification, of chromosomal DNA from a cloned U 937 transfectant which contains the expression vector pSXK1 integrated into chromosomal DNA. The intact nature of the promoter/insert sequence is indicated by the fragment size of the PCR products and the hybridisation with an HIV gene probe, which does not contain the primer sequence of the PCR products.
S 10 1 Control sample vector, 5'-junction, 887 base pair fragment 2 Control sample vector, 3'-junction, 775 base pair fragment 3 Chromosomal DNA, 5'-junction, 887 base pair fragment 4 Chromosomal DNA, 3'-junction, 775 base pair fragment Further details are given in Example 9.
Figure 8 Detection of the HIV-1 gene fragment in chromosomal DNA from cloned HUT 78 transfectants by means of PCR amplifi- O 20 cation. The PCR reaction was separated on an agarose-TAE gel and photographed under UV light following ethidium bromide staining. The PCR product contains 561 bp and corresponds to the HIV-1 gene fragment (nucleotides 699- 1305), which is also a component of the pSXK1 vector. The V 561 bp-long PCR product is indicated by an arrow.
Le A 28 518 3 Le A 28 518 37 o o 1 M DNA length standard, 516 bp and 1018 bp are indicated 1 negative control, no DNA was added to the PCR reaction 2 chromosomal DNA from U937 cells (wild type) 3 chromosomal DNA from HUT 78 cells (wild type) 4 negative control, no DNA was added to the PCR reaction positive control 6 chromosomal DNA from HUT 78 transformants (SSHUT 6/1 107-9/1) 7 chromosomal DNA from HUT 78 transformants (SSHUT 6/1 107-9/1) 8 chromosomal DNA from HUT 78 transformants (SSHUT 6/1 107-9/1) 9 chromosomal DNA from HUT 78 transformants (SSHUT 6/1 107-9/1) Figure 9 Detection of the HIV-1 gene fragment (nucleotides 4158- 5792) in chromosomal DNA from U937 monocyte cells and HUT 78 T-lymphocyte cells by means of PCR amplification.
The cells had previously been transfected with the pSXK2 vector. The PCR reaction was separated on agarose-TAE gel and photographed under UV light following ethidium bromide staining. The PCR product contains the whole HIV-1 gene fragment in pSXK2 (nucleotides 4158-5792) and is 1811 bp long. The 1811 bp-long PCR product is
D
e a .o o e a Le A 28 518 38 o 1 .1 indicated by an arrow.
DNA length standard, 1635 bp and 2036 bp length standards are indicated 1 negative control, no DNA was added to the PCR reaction 2 chromosomal DNA from U937 cells (wild type) 3 chromosomal DNA from HUT 78 cells (wild type) 4 chromosomal DNA from HUT 78 transformants (SSHUT 2/1 107-12/1) 5 chromosomal DNA from U937 transformants (SSUK 3/1 107-12/1) Figure Northern blot analysis for demonstrating several antisense RNA transcripts in a transfected, cloned U 937 cell line, which contains the expression vector KTX 11 in the cell line SSUK 20/15-KTX11b. Using a gene probe which corresponds to the HIV-1 insert DNA of the vector KTX 11, at least three antisense-RNA transcripts of 800 bp, 1,200 bp and 3,200 bp in length can be detected in the RNA of the virus-resistant cell line (track Track 2 contains RNA from the non-transfected cell line U 937 (negative control). Track 3 contains RNA molecular weight standards.
o *e *ft *o *e *e e *o oo* Figure 11 Course of HIV-1 antigen formation over 60 days following the infection (on day zero) of HUT 78 cell lines with HIV-1 LAV/BRU isolate. The optical density at 450 nm (OD 450 nm) resulting from the HIV ELISA test carried out on the cell culture supernatant is given on the ordinate. At an optical density 2.0, the culture supernatant was diluted and the indicated OD value was calculated from the dilution. The cut-off value represents the detection limit for the HIV antigen determination.
In the antisense RNA-expressing cell lines SSHUT K16/2- SXK1 and SSHUT K1/1-SXK2, HIV replication is completely inhibited over a time period of 60 days.
In the cell line HUT K14-KTX11, onset of HIV replication is delayed as compared with the non-transfected wild-type cell line HUT 78.
Figure 12 Course of HIV-1 antigen formation over 60 days following infection (on day zero) of U937 cell lines with HIV-1 LAV/BRU isolate. The optical density at 450 nm (OD 450 nm) resulting from the HIV ELISA test on the cell culture supernatant is given on the ordinate. At an optical density 2.0, the culture supernatant was d.4luted and the indicated OD value was calculated from 25 the dilution. The cut-off value represents the detection co* o*
I
limit for the HIV antigen determination.
In the antisense RNA-expressing cell line SSUK3/1-SXK2, HIV replication is completely inhibited over a time period of 60 days. The other transfected cell lines exhibit delayed HIV replication in comparison with the wild-type cell line (U937).
Figure 13 PCR analysis for detecting HIV-1 DNA in cell lines which contain antisense-RNA expression vectors and which were analysed 60 days after HIV infection. The ethidium bromide-stained agarose electrophoresis gel shows the PCR products from the chromosomal DNA of the cell lines indicated below.
A 702 base pair-long PCR fragment from the TAT-ENV gene region was amplified in HIV-infected cell lines using the primer pair 5'-ATG GAG CCA GTA GAT CCT-3' and 5'-TCT GT TCT ACC ATG TCAT. For comparison with the PCR result, the result of the HIV antigen test for the corresponding cell culture sample is given in brackets.
Track M: Track 1: Track 2: Molecular weight standard, 600 bp is indicated HUT K14-KTX11 702 bp positive (antigen test positive).
SSHUT K16/2-SXK1 702 bp positive (antigen test negative).
Le A 28 518 41
SI
Track 3: PCR product from a parallel infection experiment to track 2 Track 4: PCR product from a parallel infection experiment to track 2 Track 5: SSHUT K1/1-SXK2 702 bp positive (antigen test negative) Track 6: PCR product from a parallel infection experiment to track Track 7: Parallel infection experiment to track Track 8: HUT 78, positive control Tracks 9-11: SSUK3/1-SXK2 cultures: no 702 bp PCR product detectable (antigen test negative) Figure 14 Course of HIV antigen formation over 30 days following infection (on day zero) of HUT 78 cell lines with HIV-2 D194 isolate. The optical density at 450 nm (OD 450 nm) resulting from the HIV ELISA test from the cell culture supernatant is given on the ordinate. At an optical density 2.0, the culture supernatant was diluted and the indicated OD value was calculated from the dilution.
The cut-off value represents the detection limit of the HIV antigen determination.
In the antisense RNA-expressing cell line SSHUT K1/ 1-SXK2, HIV replication is completely inhibited over a 25 time period of 14 days. The other transfected cell lines show delayed HIV replication in comparison with the wildtype cell line (HUT 78).
A 28 518 42 ooo* Figure Course of HIV antigen formation over 30 days following infection (on day zero) of U937 cell lines with HIV-2 D194 isolate.
The optical density at 450 nm (OD 450 nm) resulting from the HIV ELISA test from the cell culture supernatant is given on the ordinate. At an optical density 2.0, the culture supernatant was diluted and the indicated OD value was calculated from the dilution. The cut-off value represents the detection limit of the HIV antigen determination.
In the antisense RNA-expressing cell line SSHUT K3/1-SXK2 HIV replication is completely inhibited over a time period of 30 days. The other transfected cell line shows delayed HIV replication in comparison to the wild-type cell line (U937).
Le A 28 518 43

Claims (4)

1. Expression vector constructs for the expression of antisense RNA, characterised in that they contain hybrid promoter gene sequences and possess a strong constitutive promoter activity or a promoter activity which can be induced in the case of attack by pathogens.
2. Expression vector constructs according to Claim 1, characterised in that they contain subgenomic gene sequences of a pathogenic virus in the orien- tation (antisense orientation) under the control of the promoter for the expression of antisense RNA, which represents a complementary nucleotide-base sequence to the mRNA of the pathogenic virus.
3. Expression vector constructs according to claims 1 and 2, characterised in that they contain proviral, subgenomic DNA in the antisense orientation from retroviruses such as HIV-1 or HIV-2 or from onco- viruses such as HTLV-1, HTLV-II or from oncogenes such as abl, erb A, erb B, fms, fos, myb, myc, ras, sis and src.
4. Expression vector constructs for the expression of antisense RNA, characterised in that they contain a promotor or a hybrid promoter gene sequences and HIV-1 proviral DNA in the antisense orientation of the nucleotides 7514-474 or 5786-474 or 2096-474 or
3825-681 or 5786-4158 or 5746-4648 or 2369-1635 or 3227-3003 or 2099-678 or 3829-2099 or 4647-4157 or S Le A 28 518 44 B *e 45
4157-3830. Expression vector constructs according to Claims 1 to 3, characterised in that they contain subgenic proviral gene sequences or oncogene sequences in the orientation for the expression of shorter antisense-RNA transcripts. 6. Expression vector constructs according to Claim 4, containing proviral gene sequences from HIV-1 of the nucleotides 605-455 or 670-440 or 825-535 or
6070-5800 or 5640-5020 or 5600+5040 or 8690-8300 or *9160-8800. c *O 7. Expression vector constructs according to Claims 1 to 6, characterised in that they contain two, three 1 or more subgenomic gene sequences from one virus or from a plurality of viruses. 8. Transfected cells which contain expression vector constructs of Claims 1 to 7. 9. Medicaments containing one or more expression vector constructs from Clams 1-7. oe00: 10. Expression vector constructs substantially as herein described with reference to any one or more of the Examples and/or the drawings. DATED this 28th day of July, 1994. BAYER AKTIENGESELLSCHAFT By Its Patent Attorneys DAVIES COLLISON CAVE Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications Abstract The present invention relates to the targeted blocking of the genetic information of the viral messenger ribo- nucleic acid (mRNA) for virus replication of HIV) in transfected, haematopoietic cells with particularly suitable expression systems, which express a comple- mentary "antisense RNA". o Le A28 518 _FC
AU29756/92A 1991-08-10 1992-11-30 Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications Ceased AU653656B2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
DE4126484A DE4126484A1 (en) 1991-08-10 1991-08-10 Anti-sense-RNA expression vectors - contain hybrid promoter sequences and viral DNA sequences i.e. HIV, in anti-sense direction, useful in prodn. of HIV-resistant cells
EP92118414A EP0594881A1 (en) 1991-08-10 1992-10-28 Expression vectors and their application in the construction of HIV-resistant human cells for therapeutic uses
CS923337A CZ284778B6 (en) 1991-08-10 1992-11-06 Constructs of expressive vectors for the expression of anti-mediator-rna, their use and medicaments containing such constructs
NZ24523092A NZ245230A (en) 1991-08-10 1992-11-23 Expression vectors coding antisense rna for targeted blocking of viral messenger rna in virus replication
CA002083812A CA2083812A1 (en) 1991-08-10 1992-11-25 Expression vectors and their use in the preparation of hiv-resistant human cells for therapeutic applications
AU29756/92A AU653656B2 (en) 1991-08-10 1992-11-30 Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications
JP4343223A JPH06153958A (en) 1991-08-10 1992-11-30 Expression vectors and use thereof in preparing hiv resisting human cell for therapeutic application

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
DE4126484A DE4126484A1 (en) 1991-08-10 1991-08-10 Anti-sense-RNA expression vectors - contain hybrid promoter sequences and viral DNA sequences i.e. HIV, in anti-sense direction, useful in prodn. of HIV-resistant cells
EP92118414A EP0594881A1 (en) 1991-08-10 1992-10-28 Expression vectors and their application in the construction of HIV-resistant human cells for therapeutic uses
CS923337A CZ284778B6 (en) 1991-08-10 1992-11-06 Constructs of expressive vectors for the expression of anti-mediator-rna, their use and medicaments containing such constructs
NZ24523092A NZ245230A (en) 1991-08-10 1992-11-23 Expression vectors coding antisense rna for targeted blocking of viral messenger rna in virus replication
CA002083812A CA2083812A1 (en) 1991-08-10 1992-11-25 Expression vectors and their use in the preparation of hiv-resistant human cells for therapeutic applications
AU29756/92A AU653656B2 (en) 1991-08-10 1992-11-30 Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications
JP4343223A JPH06153958A (en) 1991-08-10 1992-11-30 Expression vectors and use thereof in preparing hiv resisting human cell for therapeutic application

Publications (2)

Publication Number Publication Date
AU2975692A AU2975692A (en) 1994-06-09
AU653656B2 true AU653656B2 (en) 1994-10-06

Family

ID=27560534

Family Applications (1)

Application Number Title Priority Date Filing Date
AU29756/92A Ceased AU653656B2 (en) 1991-08-10 1992-11-30 Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications

Country Status (7)

Country Link
EP (1) EP0594881A1 (en)
JP (1) JPH06153958A (en)
AU (1) AU653656B2 (en)
CA (1) CA2083812A1 (en)
CZ (1) CZ284778B6 (en)
DE (1) DE4126484A1 (en)
NZ (1) NZ245230A (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6245560B1 (en) * 1990-01-18 2001-06-12 The United States Of America As Represented By The Department Of Health And Human Services Vector with multiple target response elements affecting gene expression
DE4126484A1 (en) * 1991-08-10 1993-02-11 Bayer Ag Anti-sense-RNA expression vectors - contain hybrid promoter sequences and viral DNA sequences i.e. HIV, in anti-sense direction, useful in prodn. of HIV-resistant cells
GB9125623D0 (en) * 1991-12-02 1992-01-29 Dynal As Cell modification
EP0598935A1 (en) * 1992-11-24 1994-06-01 Bayer Ag Expression vectors and their use to produce HIV-resistant human cells for therapeutic use
US5583035A (en) * 1992-12-07 1996-12-10 Bayer Aktiengesellschaft HIV antisense expression vectors
US6426412B1 (en) * 1995-12-20 2002-07-30 Subsidiary No. 3, Inc. Nucleic acids encoding human immunodeficiency virus type 1 genetic suppressor elements
US6316210B1 (en) 1995-12-20 2001-11-13 Subsidiary No. 3, Inc. Genetic suppressor elements against human immunodeficiency virus
US6071743A (en) 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
US6613506B1 (en) 2000-11-28 2003-09-02 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA1327171C (en) * 1987-06-26 1994-02-22 Lisa J. Hock Recombinant human cytomegalovirus containing foreign gene and use thereof
CA1339413C (en) * 1988-06-24 1997-09-02 Junichi Miyazaki Exogenous gene expression vector containing chick .beta.-actin gene promoter
EP0575518A1 (en) * 1991-03-06 1993-12-29 Board Of Regents, The University Of Texas System Methods and compositions for the selective inhibition of gene expression
CA2107789A1 (en) * 1991-04-05 1992-10-06 Thomas E. Wagner Retrovirus inhibition with antisense nucleic acids complementary to packaging sequences
DE4126484A1 (en) * 1991-08-10 1993-02-11 Bayer Ag Anti-sense-RNA expression vectors - contain hybrid promoter sequences and viral DNA sequences i.e. HIV, in anti-sense direction, useful in prodn. of HIV-resistant cells

Also Published As

Publication number Publication date
CZ333792A3 (en) 1994-06-15
EP0594881A1 (en) 1994-05-04
CA2083812A1 (en) 1994-05-26
AU2975692A (en) 1994-06-09
JPH06153958A (en) 1994-06-03
CZ284778B6 (en) 1999-02-17
NZ245230A (en) 1994-07-26
DE4126484A1 (en) 1993-02-11

Similar Documents

Publication Publication Date Title
Hart et al. Human chromosome 12 is required for elevated HIV-1 expression in human-hamster hybrid cells
Newstein et al. Human chromosome 12 encodes a species-specific factor which increases human immunodeficiency virus type 1 tat-mediated trans activation in rodent cells
BouHamdan et al. Human immunodeficiency virus type 1 Vpr protein binds to the uracil DNA glycosylase DNA repair enzyme
Ruben et al. Structural and functional characterization of human immunodeficiency virus tat protein
Felber et al. A quantitative bioassay for HIV-1 based on trans-activation
US8735145B2 (en) Property effecting and/or property exhibiting compositions for therapeutic and diagnostic uses
AU678157B2 (en) Method of eliminating inhibitory/instability regions of mRNA
US6114141A (en) Methods to express genes from viral vectors
Mermer et al. Identification of trans-dominant HIV-1 rev protein mutants by direct transfer of bacterially produced proteins into human cells
Ohtani et al. Electroporation: application to human lympboid cell lines for stable introduction of a transactivator gene of human T-cell leukemia virus type I
US5880276A (en) Purified retroviral constitutive transport enhancer elements that enhance nucleocytoplasmic transport of mRNA, and methods of making and using the elements
Harrison et al. Activation of a diphtheria toxin A gene by expression of human immunodeficiency virus-1 Tat and Rev proteins in transfected cells
Bar-Shira et al. An RNA secondary structure juxtaposes two remote genetic signals for human T-cell leukemia virus type I RNA 3'-end processing
AU653656B2 (en) Expression vectors and their use in the preparation of HIV-resistant human cells for therapeutic applications
US5583035A (en) HIV antisense expression vectors
Inouye et al. Potent inhibition of human immunodeficiency virus type 1 in primary T cells and alveolar macrophages by a combination anti-Rev strategy delivered in an adeno-associated virus vector
AU767620B2 (en) Conditionally replicating viral vectors and their use
Cupelli et al. Transcriptional initiation and postinitiation effects of murine leukemia virus long terminal repeat R-region sequences
EP0598935A1 (en) Expression vectors and their use to produce HIV-resistant human cells for therapeutic use
JP2001502884A (en) Inhibition of HIV-1 replication by antisense RNA expression
Schoborg et al. Definition of the RRE binding and activation domains of the caprine arthritis encephalitis virus Rev protein
Hamaia et al. The human T-cell leukemia virus type 1 Rex regulatory protein exhibits an impaired functionality in human lymphoblastoid Jurkat T cells
PL175143B1 (en) Expression vectors and their application for generation of human cells resistant against hiv for therapeutic purposes
Shimada et al. Trial of antisense RNA inhibition of HIV replication and gene expression
HU214684B (en) Method for the preparation of expression vectors and pharmaceutical compositions containing them

Legal Events

Date Code Title Description
MK14 Patent ceased section 143(a) (annual fees not paid) or expired