WO1989012461A1 - Antagonists of viral transactivating proteins - Google Patents

Antagonists of viral transactivating proteins Download PDF

Info

Publication number
WO1989012461A1
WO1989012461A1 PCT/US1989/002404 US8902404W WO8912461A1 WO 1989012461 A1 WO1989012461 A1 WO 1989012461A1 US 8902404 W US8902404 W US 8902404W WO 8912461 A1 WO8912461 A1 WO 8912461A1
Authority
WO
WIPO (PCT)
Prior art keywords
tat
peptide
virus
antagonists
peptides
Prior art date
Application number
PCT/US1989/002404
Other languages
French (fr)
Inventor
Maurice Green
Paul M. Loewenstein
Original Assignee
St. Louis University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by St. Louis University filed Critical St. Louis University
Publication of WO1989012461A1 publication Critical patent/WO1989012461A1/en

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/10011Adenoviridae
    • C12N2710/10311Mastadenovirus, e.g. human or simian adenoviruses
    • C12N2710/10322New 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
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/20011Papillomaviridae
    • C12N2710/20022New 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

Definitions

  • the present invention relates to the fields of virology and biochemistry. More particularly, the present invention relates to a method for preparing peptide inhibitors of viral gene expression and viral replication.
  • Many prominent pathogenic and cancer-inducing viruses of animal and man encode genes whose protein pro ⁇ ducts function to transcriptionally activate the expression of specific viral and/or cellular genes.
  • the evolution of viral genes which encode transactivation proteins may repre- sent a universal phenomenon that has developed to facili ⁇ tate the replication of pathogenic viruses in otherwise hostile cellular environments.
  • viral trans ⁇ activating proteins whose function is important for viral gene expression and viral replication include those encoded by the ubiquitious human herpes simplex virus (O'Hare and Hayward, 1985), the cause of numerous pathologies including the common cold sore, shingles, chicken pox and genital herpes lesions; human T cell lymphotropic virus types I (HTLVI), the etiological agent of adult T cell leukemia and HTLV2 (Chen et al., 1985; Felber et al., 1985; Sodroski et al., 1985a; Seiki et al., 1986; Fujisawa et al., 1986); human papillomavirus type 16, which is etiologically asso ⁇ ciated with cervical cancer (Phelps and Howley, 1987; Phelps et al., 1988); other serotypes of human papilloma- virus which cause sexually transmitted condylomas (Hirochika et al., 1987); human T
  • HIV human immunodeficiency virus type 1
  • AIDS acquired immune deficiency syndrome
  • the HIV tat gene product is a transactivating protein that is essential for viral gene expression and for replication of the AIDS virus (for review, see Chen, 1986) .
  • the present invention provides a method for preparing peptide antagonists of viral transactivating proteins.
  • the pep- tides are useful as therapeutic agents in the control of viral infection and viral pathogenicity.
  • FIG. 1 illustrates a flowsheet of the steps employed in the practice of the present invention.
  • Figure 2(a) illustrates that no transactivation is detected in HeLa cells microinjected with pHIV-LTRCAT alone.
  • Figure 2(b) shows that efficient transactivation is detected in HeLa cells microinjected with both plasmid pCV-1 (ptat) and pHIV-LTRCAT.
  • FIG 3 shows the amino acid sequence of the transactivating protein (tat-86) of HIV (BRU isolate).
  • Figure 4 shows a comparison of transactivation response which occurs upon coinjection of HIV-LTRCAT with ptat or with the tat-86 peptide.
  • Figure 5 represents the microscopic scoring of tat-transactivated cells.
  • Cells injected with pHIV-LTRCAT and treated with tat-86 yield mainly high grain density cells as shown by autoradiographic analysis (3+ and 4+) , whereas injected cells treated with deletion mutants (tat- dm37-72, tat-dm37-62) or with very low levels of tat-86 may show a significant proportion of 1+ and 2+ cells.
  • Panel A shows phase contrast microphotographs of representative 1+, 2+, 3+.and 4+ cells at 200 X magnification.
  • Panel B shows the same cells viewed at 200 X magnification, but out-of- phase to more clearly demonstrate the 1+ and 2+ cells.
  • Figure 6 shows the amino acid sequence of HIV-1 tat protein (BRU isolate) . Superimposed on the sequence are tentative functional regions I to IV. The absolute boundaries of these regions are not known; they are drawn for purposes of discussion and based on data of the inventors.
  • Figure 7 represents the inhibition of HIV tat- induced transactivation of the HIV LTR-driven CAT gene by chemically synthesized HIV tat deletion and amino acid substitution mutant peptides.
  • HeLa cells on coverslips were microinjected with pHIV-LTRCAT.
  • Figure 8 illustrates the transcriptional activa ⁇ tion of the E2 gene by adenovirus 5 E1A region 3 (Lillie, Loewenstein, Green, and Green, 1987) .
  • HeLa cells were microinjected with the E2 gene (in pE2) and E1A peptide or plasmid, as specified below, and then analyzed by immuno- fluorescence 18 to 24 hr. after injection with an E2- specific antibody.
  • the photographs show typical immuno- fluorescence patterns but represent only a small area of the total quadrant and therefore are not accurate quanti ⁇ tative representations.
  • Panel A pE2 - * - pm975 (encodes 289 amino acid ElA protein).
  • Panel B pE2 plus region 3 peptide.
  • Panel C pE2 plus region 1 peptide.
  • Panel D pE2 plus region 3 peptide preincubated with antibody specific to the N-terminus of region 3.
  • Figure 9 represents the sequence and localization of adenovirus 5 functional protein domains. The location of domains which encode cell DNA induction, transcriptional repression, transcriptional activation, and immortalization functions are schematically illustrated (see Lillie, Green and Green, 1986; Lillie, Loewenstein, Green and Green, 1987) .
  • Figure 10(a) shows conserved E5 regions among animal papillomavirus including reindeer papillomavirus (RPV), the deer papillomavirus (DPV), European elk papillo ⁇ mavirus (EEPV) and the bovine papillomavirus (BPVI) .
  • RSV reindeer papillomavirus
  • DDV deer papillomavirus
  • EEPV European elk papillo ⁇ mavirus
  • BPVI bovine papillomavirus
  • Figure 10(b) shows the conserved E5 region between human papillomavirus type 16 (HPV16), human papillomavirus type 18 (HPV18), and human papillomavirus type 6B (HPV6B) .
  • Figure 11(a) shows the amino acid similarity between the adenovirus type 5 ElA 13S product and the human papillomavirus type 16 E7 oncoprotein (from Phelps et al., 1988) .
  • Figure 11(b) illustrates the cellular DNA induc- tion of quiescent NIH/3T3 cells by human papillomavirus type 16 E7 deletion mutant peptides (Rawls, Pusztai, and Green, unpublished data) .
  • Figure 11(c) illustrates transactivation of the adenovirus type 5 E2 promoter by human papillomavirus HPV16 E7 deletion mutant peptides (Rawls, Pusztai, and Green, unpublished data) .
  • the present invention provides a method for pre- paring antagonists of viral transactivating proteins.
  • the terra "transactivating protein" includes oncogene products which have transactivat ⁇ ing function.
  • the present invention also provides compositions comprising the antagonists which com- positions are useful in treating an individual suffering from viral infection.
  • the invention also con ⁇ templates a method for treating an individual suffering from a viral infection by administering an effective amount of an antagonist of the transactivating protein of said virus.
  • Viral genes that encode transactivating proteins are defined by their ability to transcriptionally activate other viral genes. Putative transactivating genes are first identified by mutational analysis showing that the expres ⁇ sion of a specific viral gene is required for the subsequent expression of another viral gene or genes.
  • the viral gene encodes a transactivating protein is obtained by showing that cotransfection with a plasmid containing the gene and a plasmid containing the target promoter results in stimulation of transcription from the target.
  • the amino acid sequence of the viral transactivat ⁇ ing protein is derived from the DNA sequence by the identi ⁇ fication of open translational reading frames. This method ⁇ ology is well proven and has been employed for the identifi ⁇ cation of the transactivating proteins of essentially all viruses listed above.
  • the method of the present invention employs the following steps after the identification of a viral transactivating protein (see Figure 1).
  • An active domain of the transactivating protein is determined. This determination can be accomplished by making N-terminal and C-terminal deletions of the transactivating protein and assaying the truncated proteins for transactivating activity. Although not critical, in most cases it is preferred that one obtain the minimal active domain thereby minimizing the size of the antagonist peptide. Amino acid substitution is then made in an active domain, preferably for conserved amino acid residues, to deactivate the trans ⁇ activating function of the peptide while not destroying its binding ability thereby producing an antagonist of the viral transactivating protein.
  • the peptides of the present invention are used as therapeutic agents in the treatment of disease states asso ⁇ ciated with virus infection.
  • the peptides may be adminis ⁇ tered as free peptides or pharmaceutically acceptable salts thereof.
  • pharmaceutically acceptable salts refers to those acid addition salts or metal complexes of the peptides which do not significantly or adversely affect the therapeutic properties (e.g. efficacy, toxicity, etc.) of the peptides.
  • the peptides should be administered to individuals as a pharmaceutical composition which, in most cases, will comprise the peptide and/or pharmaceutical salts thereof with a pharmaceutically acceptable carrier.
  • compositions containing peptides of the present invention may be administered to individuals, particularly humans, either intravenously, subcutaneously, intramuscularly, intranasally or even orally. The necessary dosage will vary with the particular condition being treated, method of administration and rate of clearance of the peptide from the body.
  • peptides may be prepared by any suitable synthesis method.
  • Exemplary synthesis methods include solid-phase synthesis techniques as described in the textbook entitled “Solid-Phase Peptide Synthesis", Stewart & Young, Pierce Chemical Company, Rockford, Illinois (1984), solution synthesis and the fragment condensation synthesis methods.
  • Those skilled in the art of biochemical synthesis will recognize that such synthesis methods require the use of a protecting group to stabilize a labile side chain to prevent the side chain from being chemically altered during the synthesis process. Protection of the alpha-amino group is most commonly required to insure proper peptide bond formation, followed by selective removal of the alpha-amino protecting group to permit subsequent peptide bond formation at that location.
  • the protecting group should be stable to the reagents and conditions employed for removal of the alpha-amino protecting group at each step in the synthesis process and must be removable upon completion of the synthesis process under reaction conditions which will not detrimentally alter the peptide.
  • such peptides can be prepared using recombinant DNA or synthesis techniques.
  • genes expressing peptide antago ⁇ nists discovered by the general methodology of Fig. 1 can be cloned into suitable viral vectors (e.g. retrovirus or adenovirus) for therapeutic delivery to humans.
  • suitable viral vectors e.g. retrovirus or adenovirus
  • HIV tat peptide antagonists is an attractive possibility for the somatic cell gene therapy of hemopoietic stem cells of individuals infected with the AIDS virus.
  • amino acid nomenclature for the naturally occur ⁇ ring amino acids found in protein and comprising the peptide inhibitors of the present invention is as follows: alanine (Ala;A), asparagine (Asn;N) , aspartic acid (Asp;D), arginine (Arg;R) , cysteine (Cys;C), glutamic acid (Glu;E), glutamine (Gln;Q) , glycine (GlysG), histidine (His;H), isoleucine (lie;I), leucine (Leu;L), lysine (Lys;K), methionine (Met;M) , phenylalanine (Phe;F), proline (Pro;P), serine (Ser;S), threon
  • transactivating protein encoded by the HIV tat gene ( i) the transactivating protein encoded by the human adenovirus EIA gene, a model viral oncogene; and (iii) the cellular DNA synthesis-inducing protein encoded by the bovine papillomavirus type 1 (BPV1) E5 oncogene, and the cellular DNA synthesis-inducing and transactivating protein encoded by the human papillomavirus type 16 E7 oncogene.
  • BPV1 bovine papillomavirus type 1
  • the HIV tat transactivating protein The HIV tat transactivating protein.
  • the AIDS virus, H_ ⁇ encodes a unique transactivating protein called tat.
  • TAR transactivation response region which extends from -17 to +80 within the LTR, is required for tat transcription and contains striking inverted and direct repeats (Rosen et al., /0
  • Direct repeats are found often within transcrip ⁇ tional regulatory sequences such as enhancers and promoters (Maniatis et al., 1987).
  • HIV tat specifically recognizes the HIV LTR and tat transactivation is essential for HIV replication.
  • the tat system provides an attractive target for drug inter ⁇ vention in AIDS.
  • the open reading frame (ORF) has been defined by DNA sequence and mutational studies (for a review, see Chen, 1986) .
  • Tat function is essential for viral gene expression and for- the replication of the AIDS virus.
  • a therapy for AIDS can be developed based on peptide.
  • antagonists of the tat protein that block its func- tion The application of the invention to the identifica ⁇ tion and chemical synthesis of antagonistic tat peptides is described below. These and homologous peptides are useful as drugs to arrest the progress of AIDS.
  • Cultured human Hela cells are coinjected with (i) a recombinant plasmid that expresses a functional transactivating protein and (ii) a recombinant plasmid that contains the target promoter upstream of a suitable reporter gene.
  • the expres ⁇ sion of the reporter gene is measured by immunofluorescence at the level of the protein gene product, or by in situ hybridization at the level of mRNA. In situ hybridization has been found to be an extremely sensitive and reliable method for measuring transactivation with low background, especially with 35 S-labeled DNA probes. / /
  • the initial task was to develop a specific assay to measure transactivation by transactivating peptides, in this case, HIV tat peptides. This was achieved by the development of a cell microinjection assay for HIV tat function.
  • the first step is to show that the assay detects the activity of natural tat protein expressed constitutively from plasmid ⁇ CV-1 (also referred to as ptat) (Arya et al., 1985) .
  • the assay for transactivation function of tat peptides employs plasmid pHIV-LTRCAT (Okamoto and Wong- Staal, 1986) which contains HIV-LTR 5 ' of a chloramphenicol acetyltransferase gene (CAT) .
  • CAT expression measured as mRNA is dependent on the efficient initiation of transacti ⁇ vation of HIV-LTR. As described below, this can be achieved by co-microinjection into the cell nucleus of pHIV-LTRCAT with plasmid ptat or by the addition of tat peptide to the medium of microinjected cells.
  • the next step is to synthesize the largest peptide fragment that likely encodes a functional protein domain.
  • a large minimal active peptide is mapped by genetic analysis in which essential and non- essential coding regions in the ORF of the transactivating gene are delineated. This is illustrated by the adenovirus ElA 289 amino acid protein as described later.
  • the HIV tat protein of 86 amino acids is sufficiently small to permit initial chemical synthesis of the entire protein molecule as described below.
  • T_a_t peptides were directly added to HeLa cells immediately after microinjection with pHIV-LTRCAT using the following procedure.
  • HeLa cells are seeded in Dulbecco modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) on 22 mm 2 cover- slips in 35 mm plastic petri dishes.
  • DMEM Dulbecco modified Eagle's medium
  • FBS fetal bovine serum
  • the nuclei of 100 cells in quadrants etched on the coverslip are microinjected with the reporter plasmid pHIV- LTRCAT (or control pSV2CAT) via glass capillaries under constant pressure of a plastic syringe.
  • the CAT gene in pHIV-LTRCAT is under the control of the HIV LTR regulatory region and is not expressed significantly in HeLa cells unless cells are (i) coinjected with pCV-1 (a plasmid expressing the HIV tat 86 amino acid protein) or (ii) treated with biologically active tat-86 peptides or
  • the CAT gene in pSV2CAT is under the control of the simian virus 40 promoter, is expressed constitutively in HeLa cells, and thus serves as a control for nonspecific inhibition of transcription by
  • a peptide for tat transactivation function cells are microinjected with pHIV-LTRCAT (100 ug/ml), washed with DMEM, and peptide is added (10 -5 -10 -7 M) in 2 ml, of DMEM/0.1% bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • a mixture of test peptide and chemically synthesized tat-86 peptide (2X10 ⁇ 6 M) (sufficient to transactivate 30-35% of cells, i.e. about 50% of maximum) is added to cells in 2 ml of DMEM/0.1% BSA.
  • Cells are incubated at 37°C in a humidi- 20 fied incubator flushed with 5% C ⁇ and examined at 1 and 4 hr. for potential toxicity.
  • 1 ml of DMEM/30% FBS is added, and incubation con ⁇ tinued until 6 hr after peptide addition.
  • Cells are washed twice with phosphate-buffered saline (PBS) , once with 4% 25 paraformaldehyde/PBS/5 mM MgCl 2 / fixed in paraformaldehyde for 15 min., washed with 70% ethanol, and stored in 70% ethanol at 4° until used for hybridization.
  • PBS phosphate-buffered saline
  • the hybridization mixture consists of 2X SSC/10% dextran sulfate/2 mg/ml BSA/1 mg/ml sheared salmon sperm DNA/1 mg/ml E coli tRNA/50 mM dithiothreitol. Each 20 ul portion contains 4 X 10 6 dpm of 35 S-dCTP labeled CAT plasmid DNA (1-2 X 10 8 dpm/ug) labeled by the nick translation reaction. Post hybridization washings are performed by incubating coverslips at 37° for 30 min. in 50% formamide/2X SSC and for 30 min. in 50% formamide/lX SSC, followed by 3 to 4 washes at room temperature with shaking in IX SSC.
  • Cells are dehydrated by incubating sequentially for 5 min. periods in 70%, 95%, and absolute ethanol, followed by drying in air and mounting cell side up on microscope slides. Slides are dipped in Kodak NTB-2 emulsion at 42 deg, dried for 1 hr., and exposed for 1-2 days at 4 deg in a light-tight slide box containing a dessicant. Slides are developed in D-19 developer in total darkness, fixed, washed, and dried for at least 1 hr. prior to examination.
  • the next step is the synthesis of N-terminal deletion mutants to eliminate nonessential N-terminal sequences. Synthesis starting at the C-terminus (residue 86) was stopped after addition of amino acid residues 68, 52, 37 and 22, to yield deletion mutant peptides of 19, 35, 50 and 65 residues (see Table 1 below) . Tests for trans ⁇ activation function showed that the 65 and 50 residue t_a_t deletion mutants were quite active. These findings indi ⁇ cate that the N-terminal 36 amino acids are not essential for the tat transactivation function when added as peptide to cells. TABLE 1
  • Tat-dm37-62.47Ala a NH 2 -CFTTKALGIS AGRKKRRQRR RPPQGS-CONH 2
  • Tat-dm37-62.46Ala.47Ala a NH 2 -CFTTKALGIA AGRKKRRQRR RPPQGS-CONH 2
  • Tat-dm37-72.47Ala a NH 2 -CFTTKALGIS AGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH 2
  • Tat-dm37-72.46Ala a NH 2 -CFTTKALGIA YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH 2
  • Tat-dm37-72.41Ala a NH 2 -CFTTAALGIS YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH 2
  • Tat-dm37-72.46Ala NH 2 -CFTTKALGIA AGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH 2
  • Tat-86.41Ala a NH 2 -MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIS Y COOH
  • tat-86 may be divided tentatively int four functional domains (see Figure 6). These are referred t as regions I, II, III, and IV for the purpose of discussion a for use in the design of mutant peptides that will function as antagonists of HIV replication.
  • tat-86 an efficient (and clinically relevant) method for introducing tat peptides into cells is by peptide addition to the culture medium following the protocol described above.
  • a level of tat-86 that induces 50% of maximal transactivation is first established.
  • Candidate tat peptide are tested for antagonist activity by simultaneous addition with tat-86 to HeLa cells microinjected with pHIV-LTRCAT.
  • pHIV-LTRCAT As an internal control for non-specific inhibition of gene trans ⁇ cription by tat peptide derivatives, a quadrant of cells on the same coverslip is microinjected with pSV2CAT; constitutive expression of CAT under the control of the SV40 promoter is independent of tat expression.
  • tat deletion peptides containing only sequences that facilitate binding to putative target molecules could be antagonists of tat function. For example, they may bind to a critical site on the target molecule but may not possess the tat activation region. Thus, they may prevent fully active tat protein molecules from binding. Deletion mutant peptides that were inactive in the transactivation assa were tested for their ability to inhibit tat-86 function in th antagonist assay described in step 4.
  • Peptide ;fca.t---dm52-86 which contains 5 of the 8 amino acids in basic region III, was found to be a good antagonist of tat-86 function, exhibiting 40% inhibition of total cells transactivated at 5 x 10 ⁇ 6 M peptide (see Table 2 and Fig. 7).
  • Tat-dm49-57 which contains only basic region III, exhibits antagonist activity at high peptide concentration (see Table 2) .
  • Ta_t Peptide 2 x 10_ " ⁇ 5 _M 1. x J ). ⁇ 5 _M 5_ x 10_ "6 -M
  • amino acid substitution mutant peptides are designed and constructed as potential antagonist of tat-induced transactivation.
  • the amino acid sequence of t minimal active domain is compared by computer search with th analogous sequence in all known virus strains and isolates. I this manner, conserved amino acid residues are identified. 2 ⁇
  • the mutation of conserved residues would likely yield a biologically inactive peptide that could serve as an antagonist.
  • the mutation does not reside in a critical binding site for the target molecule but instead at a site that is still vital for function, e.g., an activation region.
  • the inventors have synthesized a series of such mutant peptides substituting alanine for individual amino acid specified in Table 1. Twenty five amino acid substitution mutant peptides built into tat minimal domain peptides or longer peptides were tested for their ability to transactivate HIVLTRCAT by the standard assay procedure. All tat peptides showing deficiency in transactivation, as detailed in Table 3, are potential antagonists. Using this approach, the inventors have identified 14 different HIV tat mutant peptides including several full length tat peptides that exhibit high levels of antagonist activity. These are described in the following sections.
  • Strong Tat antagonists are produced by substituting alanin for region II amino acids in peptide backbone tat-dm37-62. Th inventors first substituted alanine for conserved amino acids within the minimal domain peptide tat-dm37-62. In particular, tat peptides with substitutions in amino acids 40 to 47 (regio II, putative activation domain, Figure 6) that are defective i transactivation function (Table 2) were investigated. Tat-dm37 62,41Ala, iai-dm37-62,46Ala, tat-dm37-62.47Ala. and the doubl mutant £&£-dm37-62,Ala46,Ala47 are strong antagonists (Table 4, see Fig. 7). Several of these antagonists reduced wild type tat transactivation activity by 80-90%. Of particular significance, cells expressing large amounts of HIVLTR-directe CAT RNA, i.e., "high density cells,” were also reduced by 90%.
  • Effective tat antagonists are produced by substituting alanine for region II amino acids in peptide backbone tat- dm37-72. Next the same four tat amino acid substitutions were generated within tat-dm37-72 backbone to determine whether antagonist activity could be increased in this manner. Since the native 37-72 backbone possesses stronger transactivation activity than the native 37-62 backbone, it was considered tha 37-72 may be more effectively taken up by cells or more tightl bound to its target. Consistent with this possibility, all four antagonists were effective, with several showing stronger activity in the 37-72 backbone compared to the same mutant peptides in the 37-62 backbone (see Table 5) .
  • Tat antagonists in peptide backbone tat-dm37-86 containing alanine substituting for region II amino acids are also effective. Two of the substitutions that represent sites for strong antagonist activity, 41Ala and 47Ala, were built into the tat-dm37-86 backbone. Both of these peptides retain good antagonist activity (see Table 6) .
  • the data are the mean of two experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10 -6 M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total cell response and 30% high density response. The control values within each experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response. A °/
  • Tat antagonists in full length tat-86 backbone containing alanine substituting for region II amino acids were tested in the full-length tat-86 peptide backbone.
  • Tat-86,41Ala is a strong antagonist (see Table 7).
  • Tat-86,47Ala is leaky, i.e. it possesses weak transactivation activity, and accordingly its antagonist activity is diminished, but significant; presumably, the protein conformation imposed by the addition of N-terminal region I alters the activity of region II.
  • tat-86,41Ala is a potent inhibitor of wild-type tat-86 and is apparently stable over the extended period of this assay (48 hrs).
  • £iLt-86,4lAla blocks transactivation 99% at a 4-fold molar excess over that of tat-86.
  • the inventors predict that this particular analog, those listed in Table 1, part 4, and their derivatives will be potent inhibitors of HIV-1 replication.
  • Tat Peptide 2 x 1Q ⁇ 5 M 1 x 10 ⁇ 5 M 5_ x 10 ⁇ 6 M
  • the data are the mean of two experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10 ⁇ 6 M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total cell response and 30% high density response. The control values within each experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response.
  • tat antagonist peptides may produce superior antagonists.
  • Gene therapy approach for the delivery of tat antagonist peptides to HIV infected cells The inventors have synthesized full-length genes encoding some tat peptide antagonists. These mutant tat genes express tat peptides that have been shown to effectively block the function of tat-86. The inventors believe and predict that the expression of these tat mutant genes in cells will result in antagonism of HIV tat function. Such genes therefore are believed to be of value for the gene therapy of AIDS.. The inventors have cloned these tat mutant genes into several viral expression vectors. Transduction into hematopoietic stem cells of AIDS patients is a promising therapy and may avoid problems of drug delivery and drug stability.
  • the mutant peptides may be useful therapeutically for (1) the inhibition of viral infection in human beings, (2) the regul tion of human gene expression for the possible control of cancer and genetic diseases of man.
  • in vivo and vitro assays have been developed to functionally assay PD3 mutant peptides as possible antagonists of transcriptional activation.
  • preliminary data suggests that three o the mutant PD3 peptides are antagonists.
  • the application of the method of the present invention to the identification an chemical synthesis of antagonistic peptides to PD3 is describ below.
  • Step 1 The Development of Sensitive Assays for Biological Activity
  • adeno ⁇ virus ElA PD3 peptides To measure the transactivation function of adeno ⁇ virus ElA PD3 peptides, the inventors have developed two methods of assay which are highly sensitive. First, is an in vivo assay in which a target ElA-inducible gene, early adenovirus gene E2 (present in plasmid pE2) , is co-microin- jected with PD3 peptide or PD3 peptide mutants into HeLa or other suitable cell lines. Transactivation is measured 8 to 18 hours later by immuno-fluorescence which detects the E2 gene product (Lillie et al., 1987) or by in situ hybrid ⁇ ization which detects viral mRNA. Figure 8 shows the transcriptional activation of the E2 gene by ElA region 3 (Lillie., Loewenstein, Green and Green, 1987) .
  • the ElA 289 ElA protein that functions as a transactivator encodes three functional domains, PD1, PD2, and PD3 (see Figure 9) (Lillie et al., 1986; 1987); only PD3 is essen ⁇ tial for the transactivation function of the ElA oncogene. Peptides encoding the entire PD3, PD2 and PD1 domains were prepared. It is now known that PD3 alone (49 amino acids, see Table 8) is an autonomous transactivator of early viral genes both in vivo and in vitro (Lillie et al., 1987) .
  • N-terminal deletion mutants of PD3 were prepared (Table 8, dml to dm9) and tested for in vivo 7 and in vitro transactivation function. All N-terminal deletion mutants exhibited less than full activity by in vivo assay. All activity was lost after deletion of 13 N- terminal amino acids. These mutant peptides are candi- dates for ElA trans- activation antagonists. Three C- terminal deletion mutants have been synthesized (see Table 8).
  • Step 4 Development of Biological Assays to Measure Peptide Antagonist Function
  • an efficient method of introducing PD3 peptides into cells is by co-microinjec ⁇ tion with the target plasmid encoding an ElA inducible gene.
  • In vivo assays for antagonism involve coinjecting PD3 at a con- centration that exhibits half maximal activity with potential peptide antagonists and the reporter plasmid. Measurement of antagonist activity is performed by assaying the target gene product as described above.
  • In vitro assays for antagonist function involve direct competition between PD3 and potential antagonists using the standard cell-free transcription assay. The level of antagonism is determined by comparison with control in vitro assays using PD3 alone.
  • Step 5 Identification of Peptide Antagonists among Deletion Mutant Peptides that are Inactive or Possess Low Activity in the Transactivation Assay As mentioned above, several deletion mutant pep ⁇ tides have been identified that are defective in transacti ⁇ vation function. These peptides are, therefore, candidate ElA transactivation antagonists. Step 6 Further Development of Peptide Antagonists by Chemical Synthesis of Amino Acid Substitution Mutant Peptides in Minimal Domains
  • the method of this invention has been applied to the bovine papillomavirus type 1 E5 gene (Green and Loewenstein, 1987) .
  • This system is important because E5 is an oncogene and is involved in regulating cellular genes involved in cellular DNA replication.
  • E5 is an oncogene and is involved in regulating cellular genes involved in cellular DNA replication.
  • a large number of E5, peptides (Table 9 below), some of which represent potential antagonistic peptides of the bovine papillomavirus (BPV1) oncogene E5 have been prepared. It has been demonstrated that the chemically synthesized parent E5 peptide of 44 amino acids and several N-terminal deletion mutant peptides function as autonomous inducers of cellular DNA replication in growth-arrested cells (Green and Loewenstein, 1987) .
  • peptides may be useful therapeutically for the regulation of human DNA replication, and thus may be of importance for the control of some neo- plastic and genetic diseases of man.
  • the method of the present invention is applied to the bovine papillomavirus E5 protein as described below. 31
  • Step 1 The Development of Sensitive Assays for Biological Activity
  • mouse C127 cells are growth arrested by serum deprivation and then microinjected with E5 peptide preparations using glass capillaries.
  • the induction of DNA synthesis is determined by the incorporation of [ 3 H]thymidine, as measured by auto- radiography (Green and Loewenstein, 1987) .
  • a series of 14 N-terminal deletion mutants of BPV1 E5 were chemically synthesized (see Table 9) and tested as described above for DNA-synthesis induction function.
  • the C-terminal 13 amino acid peptide was found to be active.
  • the C-terminal 10 and 12 amino acid peptides were devoid of activity. These represent potential E5 antagonists.
  • the sequential addition of 7 amino acids to the C-terminal end substantially increased the DNA induction activity of the resulting peptides.
  • Activity of the C- terminal 20 amino acid peptide (E5-dm 25-44), (see Table 9) was essentially that of the entire 44 amino acid E5 protein.
  • the inventors have shown that an efficient method of introducing E5 peptides into growth arrested cells is by microinjection.
  • Assays for antagonism involve co-injecting the E5 44 amino acid peptide at a concentration that exhibits half maximal activity together with potential E5 peptide antagonists.
  • Measurement of antagonist activity is by comparison of DNA induction by the full length E5 44-mer with that of the mixture of E5 44-mer and candidate peptide antagonist.
  • E5 is conserved among animal papillomavirus type 1 (BPV1) (see Figure 10(a)). Moreover, E5 - s—a-lso conserved among two strong candidates for causative agents of cervical carcinoma and other urogenital malignancies (human papillo- mavirus type 16 (HPV16) and HPV18) r as well as one of the causative agents of venereal condylomas and juvenile laryn- geal papillomatosis (HPV6B) (see Figure 10(b)). ⁇ - /
  • E7 o ⁇ co- gene of human papillomavirus type 16 encodes a transactivation function (Phelps et al, 1988).
  • the protein encoded by E7 oncogene contains regions of remarkable similarity to those of human adenovirus ElA protein domains 1, 2, and 3 (see Figure 11(a)).
  • the inventors have chemi- cally synthesized the full E7 oncoprotein of 98 amino acids as well as a series of deletion mutant peptides. They have demonstrated that regions 1 and 2 function as an oncogene to induce cellular DNA synthesis ( Figure 11(b)). Further ⁇ more, they have shown that region 3 functions as a trans- activator (Figure 11(c)).
  • E5-dm(l-43) NH 2 -MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGLP-CONH, E5-dm(l-42) NH,-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGL-CONH 2 10
  • E5-dm(l-41) NH 2 -MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTG-CONH 2
  • E5-dm(l-40) NH 2 -MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCT-CONH 2 E5-dm(1-3,9) NH,-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSC-CONH,

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)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A method for preparing antagonists of viral transactivating proteins is disclosed. In one aspect the method involves the preparation of antagonists from active domains of the viral transactivating protein. Pharmaceutical compositions comprising the antagonists which compositions are useful in treating an individual suffering from viral infection are also disclosed.

Description

I ANTAGONISTS OF VIRAL TRANSACTIVATING PROTEINS CROSS REFERENCE TO RELATED APPLICATION
This application is a continuation-in-part of application Serial No. 207,393 filed June 16, 1988.
BACKGROUND OF THE INVENTION
The present invention relates to the fields of virology and biochemistry. More particularly, the present invention relates to a method for preparing peptide inhibitors of viral gene expression and viral replication. Many prominent pathogenic and cancer-inducing viruses of animal and man encode genes whose protein pro¬ ducts function to transcriptionally activate the expression of specific viral and/or cellular genes. The evolution of viral genes which encode transactivation proteins may repre- sent a universal phenomenon that has developed to facili¬ tate the replication of pathogenic viruses in otherwise hostile cellular environments. Examples of viral trans¬ activating proteins whose function is important for viral gene expression and viral replication include those encoded by the ubiquitious human herpes simplex virus (O'Hare and Hayward, 1985), the cause of numerous pathologies including the common cold sore, shingles, chicken pox and genital herpes lesions; human T cell lymphotropic virus types I (HTLVI), the etiological agent of adult T cell leukemia and HTLV2 (Chen et al., 1985; Felber et al., 1985; Sodroski et al., 1985a; Seiki et al., 1986; Fujisawa et al., 1986); human papillomavirus type 16, which is etiologically asso¬ ciated with cervical cancer (Phelps and Howley, 1987; Phelps et al., 1988); other serotypes of human papilloma- virus which cause sexually transmitted condylomas (Hirochika et al., 1987); human hepatitis B virus which is strongly implicated in hepatocellular carcinoma (Twu and Schloemer, 1987; Spandau and Lee, 1988); bovine leukemia virus (Rosen et al., 1985; Derse, 1987); simian virus 40 (Brady et al., 1984; Keller and Alwine, 1984); pseudorabies virus (Ihara et al., 1983) and human adenovirus types 2 and 5.
An example of urgent medical importance is human immunodeficiency virus type 1 (HIV), the primary etiological agent of acquired immune deficiency syndrome (AIDS) . The HIV tat gene product is a transactivating protein that is essential for viral gene expression and for replication of the AIDS virus (for review, see Chen, 1986) .
Evidence suggests that similar mechanisms are involved in the transactivating functions of these viral gene products (Kingston et al., 1985 and Berk, 1986). The present invention provides a method for preparing peptide antagonists of viral transactivating proteins. The pep- tides are useful as therapeutic agents in the control of viral infection and viral pathogenicity.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 illustrates a flowsheet of the steps employed in the practice of the present invention.
Figure 2(a) illustrates that no transactivation is detected in HeLa cells microinjected with pHIV-LTRCAT alone.
Figure 2(b) shows that efficient transactivation is detected in HeLa cells microinjected with both plasmid pCV-1 (ptat) and pHIV-LTRCAT.
Figure 3 shows the amino acid sequence of the transactivating protein (tat-86) of HIV (BRU isolate).
Figure 4 shows a comparison of transactivation response which occurs upon coinjection of HIV-LTRCAT with ptat or with the tat-86 peptide. Figure 5 represents the microscopic scoring of tat-transactivated cells. Cells injected with pHIV-LTRCAT and treated with tat-86 yield mainly high grain density cells as shown by autoradiographic analysis (3+ and 4+) , whereas injected cells treated with deletion mutants (tat- dm37-72, tat-dm37-62) or with very low levels of tat-86 may show a significant proportion of 1+ and 2+ cells. Panel A shows phase contrast microphotographs of representative 1+, 2+, 3+.and 4+ cells at 200 X magnification. Panel B shows the same cells viewed at 200 X magnification, but out-of- phase to more clearly demonstrate the 1+ and 2+ cells.
Figure 6 shows the amino acid sequence of HIV-1 tat protein (BRU isolate) . Superimposed on the sequence are tentative functional regions I to IV. The absolute boundaries of these regions are not known; they are drawn for purposes of discussion and based on data of the inventors.
Figure 7 represents the inhibition of HIV tat- induced transactivation of the HIV LTR-driven CAT gene by chemically synthesized HIV tat deletion and amino acid substitution mutant peptides. HeLa cells on coverslips were microinjected with pHIV-LTRCAT. To different cover¬ slips were added: (upper panel tat-86 alone (2X10_6M); (second panel from top) tat-86 (2X10~6M) + tat-dm52-86 (10_!5M); (second panel from bottom) tat-86 (2X10"6M) + £a±-dm37-62,47Ala (10_5M); (bottom panel) tat-86 (1X10"6M) + tat-dm37-62,46Ala (10_5M). Six hours after peptide addition, cells were fixed and analyzed by in situ hybridization using 35S-dCTP labeled CAT plasmid DNA. The photographs show typical autoradiographic patterns.
Figure 8 illustrates the transcriptional activa¬ tion of the E2 gene by adenovirus 5 E1A region 3 (Lillie, Loewenstein, Green, and Green, 1987) . HeLa cells were microinjected with the E2 gene (in pE2) and E1A peptide or plasmid, as specified below, and then analyzed by immuno- fluorescence 18 to 24 hr. after injection with an E2- specific antibody. The photographs show typical immuno- fluorescence patterns but represent only a small area of the total quadrant and therefore are not accurate quanti¬ tative representations. Panel A; pE2 -*- pm975 (encodes 289 amino acid ElA protein). Panel B: pE2 plus region 3 peptide. Panel C; pE2 plus region 1 peptide. Panel D; pE2 plus region 3 peptide preincubated with antibody specific to the N-terminus of region 3.
Figure 9 represents the sequence and localization of adenovirus 5 functional protein domains. The location of domains which encode cell DNA induction, transcriptional repression, transcriptional activation, and immortalization functions are schematically illustrated (see Lillie, Green and Green, 1986; Lillie, Loewenstein, Green and Green, 1987) .
Figure 10(a) shows conserved E5 regions among animal papillomavirus including reindeer papillomavirus (RPV), the deer papillomavirus (DPV), European elk papillo¬ mavirus (EEPV) and the bovine papillomavirus (BPVI) .
Figure 10(b) shows the conserved E5 region between human papillomavirus type 16 (HPV16), human papillomavirus type 18 (HPV18), and human papillomavirus type 6B (HPV6B) . Figure 11(a) shows the amino acid similarity between the adenovirus type 5 ElA 13S product and the human papillomavirus type 16 E7 oncoprotein (from Phelps et al., 1988) .
Figure 11(b) illustrates the cellular DNA induc- tion of quiescent NIH/3T3 cells by human papillomavirus type 16 E7 deletion mutant peptides (Rawls, Pusztai, and Green, unpublished data) .
Figure 11(c) illustrates transactivation of the adenovirus type 5 E2 promoter by human papillomavirus HPV16 E7 deletion mutant peptides (Rawls, Pusztai, and Green, unpublished data) .
STATEMENT OF THE INVENTION
The present invention provides a method for pre- paring antagonists of viral transactivating proteins. For purposes of the present invention, the terra "transactivating protein" includes oncogene products which have transactivat¬ ing function. In another aspect the present invention also provides compositions comprising the antagonists which com- positions are useful in treating an individual suffering from viral infection. Accordingly, the invention also con¬ templates a method for treating an individual suffering from a viral infection by administering an effective amount of an antagonist of the transactivating protein of said virus. Viral genes that encode transactivating proteins are defined by their ability to transcriptionally activate other viral genes. Putative transactivating genes are first identified by mutational analysis showing that the expres¬ sion of a specific viral gene is required for the subsequent expression of another viral gene or genes. Experimental proof that the viral gene encodes a transactivating protein is obtained by showing that cotransfection with a plasmid containing the gene and a plasmid containing the target promoter results in stimulation of transcription from the target. The amino acid sequence of the viral transactivat¬ ing protein is derived from the DNA sequence by the identi¬ fication of open translational reading frames. This method¬ ology is well proven and has been employed for the identifi¬ cation of the transactivating proteins of essentially all viruses listed above.
Briefly, the method of the present invention employs the following steps after the identification of a viral transactivating protein (see Figure 1). An active domain of the transactivating protein is determined. This determination can be accomplished by making N-terminal and C-terminal deletions of the transactivating protein and assaying the truncated proteins for transactivating activity. Although not critical, in most cases it is preferred that one obtain the minimal active domain thereby minimizing the size of the antagonist peptide. Amino acid substitution is then made in an active domain, preferably for conserved amino acid residues, to deactivate the trans¬ activating function of the peptide while not destroying its binding ability thereby producing an antagonist of the viral transactivating protein. Conserved residues may be deter¬ mined by comparing the sequence of virus from the same class. Viral sequences are contained in publicly available databases such as Genbank (Intelligenetics) and NBRF (National Biomedical Research Foundation) . It should be noted that fragments of transactivating proteins which do not have transactivating activity may possess the desired antagonistic activity. Hence, the mutation of active domains should only be considered a preferred method of obtaining peptide antagonists. The general methodology for practice of the present invention is outlined in Figure 1, and is believed to be applicable to all the viruses listed above.
The peptides of the present invention are used as therapeutic agents in the treatment of disease states asso¬ ciated with virus infection. The peptides may be adminis¬ tered as free peptides or pharmaceutically acceptable salts thereof. The term "pharmaceutically acceptable salts" refers to those acid addition salts or metal complexes of the peptides which do not significantly or adversely affect the therapeutic properties (e.g. efficacy, toxicity, etc.) of the peptides. The peptides should be administered to individuals as a pharmaceutical composition which, in most cases, will comprise the peptide and/or pharmaceutical salts thereof with a pharmaceutically acceptable carrier. The term "pharmaceutically acceptable carrier" refers to those solid and liquid carriers which do not significantly or adversely affect the therapeutic properties of the peptides. The pharmaceutical compositions containing peptides of the present invention may be administered to individuals, particularly humans, either intravenously, subcutaneously, intramuscularly, intranasally or even orally. The necessary dosage will vary with the particular condition being treated, method of administration and rate of clearance of the peptide from the body.
The above-described peptides may be prepared by any suitable synthesis method. Exemplary synthesis methods include solid-phase synthesis techniques as described in the textbook entitled "Solid-Phase Peptide Synthesis", Stewart & Young, Pierce Chemical Company, Rockford, Illinois (1984), solution synthesis and the fragment condensation synthesis methods. Those skilled in the art of biochemical synthesis will recognize that such synthesis methods require the use of a protecting group to stabilize a labile side chain to prevent the side chain from being chemically altered during the synthesis process. Protection of the alpha-amino group is most commonly required to insure proper peptide bond formation, followed by selective removal of the alpha-amino protecting group to permit subsequent peptide bond formation at that location. In selecting a particular side chain protecting group to be employed in the synthesis of such peptides, the protecting group should be stable to the reagents and conditions employed for removal of the alpha-amino protecting group at each step in the synthesis process and must be removable upon completion of the synthesis process under reaction conditions which will not detrimentally alter the peptide. For commercial- scale quantities of peptides, such peptides can be prepared using recombinant DNA or synthesis techniques.
Finally, it is recognized that by the use of recombinant DNA techniques, genes expressing peptide antago¬ nists discovered by the general methodology of Fig. 1 can be cloned into suitable viral vectors (e.g. retrovirus or adenovirus) for therapeutic delivery to humans. In par¬ ticular, this approach with HIV tat peptide antagonists is an attractive possibility for the somatic cell gene therapy of hemopoietic stem cells of individuals infected with the AIDS virus.
All peptide structures represented in the follow¬ ing description and claims are shown in conventional format wherein the amino group at the N-terminus appears to the left and the carboxyl group at the C-terminus at the right. Likewise, amino acid nomenclature for the naturally occur¬ ring amino acids found in protein and comprising the peptide inhibitors of the present invention is as follows: alanine (Ala;A), asparagine (Asn;N) , aspartic acid (Asp;D), arginine (Arg;R) , cysteine (Cys;C), glutamic acid (Glu;E), glutamine (Gln;Q) , glycine (GlysG), histidine (His;H), isoleucine (lie;I), leucine (Leu;L), lysine (Lys;K), methionine (Met;M) , phenylalanine (Phe;F), proline (Pro;P), serine (Ser;S), threonine (Thr;T), tryptophan (Trp;W), tyrosine (Tyr;Y), and valine (Val;V).
For sake of clarity and brevity in the explana¬ tion of the present invention, the description will focus on application of the invention to the following viral systems:
( i) the transactivating protein encoded by the HIV tat gene; ( ii) the transactivating protein encoded by the human adenovirus EIA gene, a model viral oncogene; and (iii) the cellular DNA synthesis-inducing protein encoded by the bovine papillomavirus type 1 (BPV1) E5 oncogene, and the cellular DNA synthesis-inducing and transactivating protein encoded by the human papillomavirus type 16 E7 oncogene. It will be understood that the description that follows is equally applicable to other viruses that encode a trans- activating gene, including the herpes simplex viruses and the human hepatitis B virus.
HUMAN IMMUNODEFICIENCY VIRUS TYPE 1 (HIV)
The HIV tat transactivating protein. The AIDS virus, H_ψ , encodes a unique transactivating protein called tat. an
Figure imgf000011_0001
1987; Peterlin et al., 1986; Rosen et al., 1986; Sodroski et al., 1985b; Wright et al., 1986). The mechanism of tat transactivation is unclear. Evidence has been presented for (i) an increase in the steady state level of HIV- specific mRNA and/or (ii) an increase in the translational utilization of that mRNA (Cullen, 1986; Wright et al., 1986) . More recently, evidence was reported that tat increases HIV mRNA levels by enhancing the rate of trans¬ cription rather than stabilizing HIV specific mRNAs (Hauber et al., 1987). The presumed target of tat action, the TAR (transactivation response) region which extends from -17 to +80 within the LTR, is required for tat transcription and contains striking inverted and direct repeats (Rosen et al., /0
1986) . Direct repeats are found often within transcrip¬ tional regulatory sequences such as enhancers and promoters (Maniatis et al., 1987).
HIV tat specifically recognizes the HIV LTR and tat transactivation is essential for HIV replication. Thus, the tat system provides an attractive target for drug inter¬ vention in AIDS. The relatively small size of the tat gene product, 86 amino acids, places it within the limits of peptide synthesis. The open reading frame (ORF) has been defined by DNA sequence and mutational studies (for a review, see Chen, 1986) . Tat function is essential for viral gene expression and for- the replication of the AIDS virus. Hence, a therapy for AIDS can be developed based on peptide. antagonists of the tat protein that block its func- tion. The application of the invention to the identifica¬ tion and chemical synthesis of antagonistic tat peptides is described below. These and homologous peptides are useful as drugs to arrest the progress of AIDS.
Step 1 Development of Sensitive Assays for Biological Activity
Cultured human Hela cells (or other suitable cultured cells) are coinjected with (i) a recombinant plasmid that expresses a functional transactivating protein and (ii) a recombinant plasmid that contains the target promoter upstream of a suitable reporter gene. The expres¬ sion of the reporter gene is measured by immunofluorescence at the level of the protein gene product, or by in situ hybridization at the level of mRNA. In situ hybridization has been found to be an extremely sensitive and reliable method for measuring transactivation with low background, especially with 35S-labeled DNA probes. / /
The initial task was to develop a specific assay to measure transactivation by transactivating peptides, in this case, HIV tat peptides. This was achieved by the development of a cell microinjection assay for HIV tat function. The first step is to show that the assay detects the activity of natural tat protein expressed constitutively from plasmid ρCV-1 (also referred to as ptat) (Arya et al., 1985) . The assay for transactivation function of tat peptides employs plasmid pHIV-LTRCAT (Okamoto and Wong- Staal, 1986) which contains HIV-LTR 5 ' of a chloramphenicol acetyltransferase gene (CAT) . CAT expression measured as mRNA is dependent on the efficient initiation of transacti¬ vation of HIV-LTR. As described below, this can be achieved by co-microinjection into the cell nucleus of pHIV-LTRCAT with plasmid ptat or by the addition of tat peptide to the medium of microinjected cells.
In the assay, HeLa cells are co-microinjected with pHIV-LTRCAT and pCV-1. The activation of the HIV LTR is measured 6 hr. later by in situ hybridization analysis that measures the expression of the CAT gene as mRNA. Referring to Figure 2(a), no detectable HIV-LTR transactivation is detected in cells microinjected with pHIV-LTRCAT alone. However, co-microinjection with pCV-1 dramatically induces HIV-LTR-driven expression of CAT, as shown by the massive cellular accumulation of CAT mRNA, visualized by the coalescence of autoradiographic grains above positive cells, see Figure 2(b). From 60 to 80% of microinjected cells are transactivated by co-microinjection. This assay for tat transactivation function is rapid, extremely sensi- tive and reliable. The assay system responds well to chemically synthesized tat peptides and deletion mutants that encode the tat transactivation domain. IZ
Step 2 Synthesis of a Large Peptide Representing the Putative Active Domain
The next step is to synthesize the largest peptide fragment that likely encodes a functional protein domain. For large proteins, a large minimal active peptide is mapped by genetic analysis in which essential and non- essential coding regions in the ORF of the transactivating gene are delineated. This is illustrated by the adenovirus ElA 289 amino acid protein as described later. The HIV tat protein of 86 amino acids is sufficiently small to permit initial chemical synthesis of the entire protein molecule as described below.
The full length tat 86 peptide was chemically synthesized by the solid-phase method of Merrifield (1963) using tBoc chemistry and a manual adaptation of the auto¬ mated procedure outlined in Clark-Lewis et al., (1986). Figure 3 shows the sequence of tat-86.
The cell microinjection/ situ hybridization assay was further developed to assay transactivation by tat peptides. T_a_t peptides were directly added to HeLa cells immediately after microinjection with pHIV-LTRCAT using the following procedure.
1. Cell microiniection and peptide treatment. HeLa cells are seeded in Dulbecco modified Eagle's medium (DMEM) containing 10% fetal bovine serum (FBS) on 22 mm2 cover- slips in 35 mm plastic petri dishes. When the cells are 50-70% confluent, the nuclei of 100 cells in quadrants etched on the coverslip (each quadrant contains 400-600 cells) are microinjected with the reporter plasmid pHIV- LTRCAT (or control pSV2CAT) via glass capillaries under constant pressure of a plastic syringe. The CAT gene in pHIV-LTRCAT is under the control of the HIV LTR regulatory region and is not expressed significantly in HeLa cells unless cells are (i) coinjected with pCV-1 (a plasmid expressing the HIV tat 86 amino acid protein) or (ii) treated with biologically active tat-86 peptides or
5 deletion tat peptides containing the active regions for the tat transactivation function. The CAT gene in pSV2CAT is under the control of the simian virus 40 promoter, is expressed constitutively in HeLa cells, and thus serves as a control for nonspecific inhibition of transcription by
10 candidate tat peptide antagonists.
To assay a peptide for tat transactivation function, cells are microinjected with pHIV-LTRCAT (100 ug/ml), washed with DMEM, and peptide is added (10-5-10-7M) in 2 ml, of DMEM/0.1% bovine serum albumin (BSA). To assay y< 15 potential antagonists of tat transactivation function, a mixture of test peptide and chemically synthesized tat-86 peptide (2X10~6M) (sufficient to transactivate 30-35% of cells, i.e. about 50% of maximum) is added to cells in 2 ml of DMEM/0.1% BSA. Cells are incubated at 37°C in a humidi- 20 fied incubator flushed with 5% Cθ and examined at 1 and 4
Figure imgf000015_0001
hr. for potential toxicity. At 2 hr. after peptide addi¬ tion, 1 ml of DMEM/30% FBS is added, and incubation con¬ tinued until 6 hr after peptide addition. Cells are washed twice with phosphate-buffered saline (PBS) , once with 4% 25 paraformaldehyde/PBS/5 mM MgCl2/ fixed in paraformaldehyde for 15 min., washed with 70% ethanol, and stored in 70% ethanol at 4° until used for hybridization.
2. In situ hybridization and autoradiographv. Coverslips are incubated at room temperature for 10 min. in PBS/5 mM 30 MgCl2 and for 10 min. in 0.1 M glycine/0.2 M Tris, pH 7.4, followed by 10 min. at 65° in 50% formamide/2X SSC (SSC = 0.15 M NaCl/0.015 M Na3 citrate). Coverslips are placed cell side down on parafilm containing 20 ul amounts of hybridization mixture and incubated at 37° for 5 to 16 hr. The hybridization mixture consists of 2X SSC/10% dextran sulfate/2 mg/ml BSA/1 mg/ml sheared salmon sperm DNA/1 mg/ml E coli tRNA/50 mM dithiothreitol. Each 20 ul portion contains 4 X 106 dpm of 35S-dCTP labeled CAT plasmid DNA (1-2 X 108 dpm/ug) labeled by the nick translation reaction. Post hybridization washings are performed by incubating coverslips at 37° for 30 min. in 50% formamide/2X SSC and for 30 min. in 50% formamide/lX SSC, followed by 3 to 4 washes at room temperature with shaking in IX SSC. Cells are dehydrated by incubating sequentially for 5 min. periods in 70%, 95%, and absolute ethanol, followed by drying in air and mounting cell side up on microscope slides. Slides are dipped in Kodak NTB-2 emulsion at 42 deg, dried for 1 hr., and exposed for 1-2 days at 4 deg in a light-tight slide box containing a dessicant. Slides are developed in D-19 developer in total darkness, fixed, washed, and dried for at least 1 hr. prior to examination.
3. Microscopic scoring of HIV tat Transactivated Cells in Autoradiographs. Experimental slides are routinely examined after autoradiography by phase microscopy at a magnification of 200X. Cells are scored from 4+ to 1+ as follows: 4+ response - the emulsion over cells is black, i.e. silver grains are packed, coalesce, and obscure the cell structure beneath the emulsion; 3+ response - silver grains are tightly packed and too numerous to count but cell structure is visible beneath the emulsion; 2+ and 1+ response - apparent at 400 X and higher magnificaion, grain counts are 100 to 200 for 2+ and about 50 to 100 per cell for 1+. Hereafter, 4+ and 3+ are referred to as "high grain density" positives whereas 2+ and 1+ are referred to as "low density." In most experiments, control HeLa cells not microinjected and cells microinjected with pHIV-LTRCAT but not exposed to ptat or tat peptides show no significant background grain counts with a 3^S-labeled CAT DNA probe. Thus, the assay is both specific and highly sensitive. Cells microinjected with pHIV-LTRCAT plus pCV-1 (see Figure 2) or microinjected with pHIV-LTRCAT and treated with high levels of active tat peptide (see Figure 4) reveal mainly 4+ and 3+ response. Cells microinjected with pHIV-LTRCAT and treated with lower level of tat peptides exhibit increasing proportions of 2+ and 1+ cells (see Figure 5) . This assay has the advantage that it mimics the clinical situation in which tat antagonist peptide would be administered to AIDS patients. The transactiva ion response by the chemically synthesized tat-86 peptide is remarkably efficient and rapid. A comparison with the transactivation response which occurs upon coinjection of HIV-LTRCAT with the tat-86 peptide is shown in Figure 4. These results demonstrate that the chemically synthesized full length tat protein effectively enters the cell nucleus and transactivates the HIV-LTRCAT. This finding provides the experimental basis for the delineation of a smaller functional peptide domain and for the development of peptide antagonists of tat.
Step 3 Identification of Smaller Peptides that Exhibit Transactivating Function bv Sequential Synthesis and Assay of N-terminal and C-Terminal Deletion Mutants
The next step is the synthesis of N-terminal deletion mutants to eliminate nonessential N-terminal sequences. Synthesis starting at the C-terminus (residue 86) was stopped after addition of amino acid residues 68, 52, 37 and 22, to yield deletion mutant peptides of 19, 35, 50 and 65 residues (see Table 1 below) . Tests for trans¬ activation function showed that the 65 and 50 residue t_a_t deletion mutants were quite active. These findings indi¬ cate that the N-terminal 36 amino acids are not essential for the tat transactivation function when added as peptide to cells. TABLE 1
Chemically Synthesized Tat-86 Peptides
1. N-TERMINAL DELETION MUTANTS
Tat-dm22-86* CTTCY CKKCCFHCQV CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ VSLSKQPTSQ-
PRGDPTGPKE-COOH
5 Tat-dm37-86* CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ VSLSKQPTSQ PRGDPTGPKE-COOH Tat-dm52-86a RRQRR RPPQGSQTHQ VSLSKQPTSQ PRGDPTGPKE-COOH Tat-dm68-86g SLSKQPTSQ PRGDPTGPKE-COOH
2. N- AND C-TERMINAL DELETION MUTANTS
Tat-dm32-57* NH2-FHCQV CFTTKALGIS YGRKKRRQRR R-CONH2 10 Tat-dm32-62* NH2-FHCQV CFTTKALGIS YGRKKRRQRR RPPQGS-CONH2 Tat-dm32-67* NH2-FHCQV CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ V-CONH2 Tat-dm32-72+ NH2-FHCQV CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ VSLSKQ-CONH2 Tat-dm37-57* NH2-CFTTKALGIS YGRKKRRQRR R-CONH2 Tat-dra37-62* NH2-CFTTKALGIS YGRKKRRQRR RPPQGS-CONH2 15 Tat-dm37-67 NH2-CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ V-CONH2 Tat-dm37-72* NH '2,-CFTTKALGIS YGRKKRRQRR RPPQGSQTHQ VSLSKQ-CONH,
Tat-dm49-57a NH.-RKKRRQRR R-CONH,
TABLE 1 (continued)
3. AMINO ACID SUBSTITUTION. N-. C-TERMINAL DELETION MUTANTS
Tat-dm37-62.54Ala* NH2-CFTTKALGIS YGRKKRRARR RPPQGS-CONH2
Tat-dm37-62.47Alaa NH2-CFTTKALGIS AGRKKRRQRR RPPQGS-CONH2
5 Tat-dm37-62.46Alaa NH2-CFTTKALGIA YGRKKRRQRR RPPQGS-CONH2
Tat-dm37-62.41Alaa NH2-CFTTAALGIS YGRKKRRQRR RPPQGS-CONH2
Tat-dm37-62,40Alac NH2-CFTAKALGIS YGRKKRRQRR RPPQGS-CONH2
Tat-dm37-62,39Alac NH2-CFATKALGIS YGRKKRRQRR RPPQGS-CONH2
Tat-dm37-62,38Ala* NH2-CATTKALGIS YGRKKRRQRR RPPQGS-CONH2
10 Tat-dm37-62.37Ala* NH2-AFTTKALGIS YGRKKRRQRR RPPQGS-CONH2 i
Tat-dm37-62.46Ala.47Alaa NH2-CFTTKALGIA AGRKKRRQRR RPPQGS-CONH2
Tat-dm37-72.47Alaa NH2-CFTTKALGIS AGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
Tat-dm37-72.46Alaa NH2-CFTTKALGIA YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
Tat-dm37-72.41Alaa NH2-CFTTAALGIS YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
15 Tat-dm37-72.40Alac NH2-CFTAKALGIS YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
Tat-dm37-72,39Alac NH2-CFATKALGIS YGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
Tat-dm37-72.46Ala. NH2-CFTTKALGIA AGRKKRRQRR RPPQGSQTHQVSLSKQ-CONH2
47Alaa
4. FULL LENGTH TAT-86 PEPTIDES
20 Tat-86 NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTKALGIS Y
GRKKRRQRR RPPQGSQTHQ VSLSKQPTSQ PRGDPTGPKE-COOH
TABLE 1 (continued)
Tat-86.22Alab NH2-MEPVDP RLEPWKHPGS QPKTAATTCY CKKCCFHCQV CFTTKALGIS Y COOHd
Tat-86.25Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTAY CKKCCFHCQV CFTTKALGIS Y COOH
Tat-86.27Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTCY AKKCCFHCQV CFTTKALGIS Y COOH
5 Tat-86.30Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTKALGIS Y COOH
Tat-86.31Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCAFHCQV CFTTKALGIS Y COOH
Tat-86.34Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTKALGIS Y COOH
Tat-86.37Alab NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV AFTTKALGIS Y COOH
Tat-86.41Alaa NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIS Y COOH
10 Tat-86.47Alaa NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTKALGIS A COOH
Tat-86.41Ala. NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIS A COOH ex.
47Alab
Tat-86,41Ala, NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIA Y COOH
46Alab
15 Ta t-86 . 46Ala . NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTKALGIA Y COOH
Tat-86 . 41Ala , NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIA Y COOH
46Alab
Tat-86 . 41Ala . NH2-MEPVDP RLEPWKHPGS QPKTACTTCY CKKCCFHCQV CFTTAALGIA A COOH
46Ala , 47Alab
, ,
20 *Actιve in HIVLTR transactivation; aAntagonist of HIVLTR transaction; bPotential antagonist under test; cDiminished activity; ddesignates tat
To establish how much of the C-terminal domain is required for tat peptide transactivation, a series of peptid starting at residues 72, 67, 62 and 57 and terminating synthe after addition of amino acid residues 37 or 32 (see Table 1) were synthesized. DNA transfection studies have indicated th tat protein lacking the second exon (residues 73-86) can stil transactivate the HIV-LTR (Muesing et al., 1987). All of th N-terminal/C-terminal deletion mutants were tested for activi and found to be active (Table 1) . Deletion of 14 amino acids from the C-terrainus (and 36 amino acids from the N-terminus) yielded a mutant peptide (tat-dm 37-72) that exhibits activit close to that of the 50 amino acid peptide, tat-dm37-86, whi possesses the native C-terminus. Deletion of 10 additional amino acids produced a mutant peptide, tat-dra37-62, that still possesses clear but reduced transactivation activity. This particular mutant peptide has proven very useful because sing amino acid substitution mutations built into this peptide backbone by chemical synthesis are strong antagonists of tat transactivation (see step 6 below) . To summarize, the inventors' findings have led the to the conclusion that tat-86 may be divided tentatively int four functional domains (see Figure 6). These are referred t as regions I, II, III, and IV for the purpose of discussion a for use in the design of mutant peptides that will function as antagonists of HIV replication.
Step 4 Development of Biological Assays to Measure Peptide Antagonist Function
The inventors have shown that an efficient (and clinically relevant) method for introducing tat peptides into cells is by peptide addition to the culture medium following the protocol described above. To assay for antagonistic function, a level of tat-86 that induces 50% of maximal transactivation is first established. Candidate tat peptide are tested for antagonist activity by simultaneous addition with tat-86 to HeLa cells microinjected with pHIV-LTRCAT. As an internal control for non-specific inhibition of gene trans¬ cription by tat peptide derivatives, a quadrant of cells on the same coverslip is microinjected with pSV2CAT; constitutive expression of CAT under the control of the SV40 promoter is independent of tat expression.
Step 5 Identification of Peptide Antagonists Among Deletion Mutant Peptides that are Inactive or Possess Low Activity in the Transactivation Assay
In principle, tat deletion peptides containing only sequences that facilitate binding to putative target molecules could be antagonists of tat function. For example, they may bind to a critical site on the target molecule but may not possess the tat activation region. Thus, they may prevent fully active tat protein molecules from binding. Deletion mutant peptides that were inactive in the transactivation assa were tested for their ability to inhibit tat-86 function in th antagonist assay described in step 4. Peptide ;fca.t---dm52-86, which contains 5 of the 8 amino acids in basic region III, was found to be a good antagonist of tat-86 function, exhibiting 40% inhibition of total cells transactivated at 5 x 10~6 M peptide (see Table 2 and Fig. 7). Tat-dm49-57, which contains only basic region III, exhibits antagonist activity at high peptide concentration (see Table 2) .
TABLE 2
Inhibition of Tat-Mediated HIVLTR Transactivation by Tat Deletion Peptides Added to Cells
Transactivated Cells % Inhibition
Total (High Density)
Ta_t Peptide 2 x 10_"~5_M 1. x J ).~5_M 5_ x 10_"6-M
52-86 60 (80) • 52 (70) 40 (34)
49-57 31 (37) 20 (0) 0 (0)
Treatment with tat-86 alone gave an average of 54% total cel response and 30% high density response. The control values within each experiment were normalized to 100% for calculatio of the percent inhibition of total cell and high density cel response.
Step 6 Further Development of Peptide Antagonists by Chemical Synthesis of Amino Acid Substitution Mutant Peptides in Minimal Domains
Once active peptide domains and preferably minimal domains are established, amino acid substitution mutant peptides are designed and constructed as potential antagonist of tat-induced transactivation. The amino acid sequence of t minimal active domain is compared by computer search with th analogous sequence in all known virus strains and isolates. I this manner, conserved amino acid residues are identified. 2Λ
Logically, the mutation of conserved residues would likely yield a biologically inactive peptide that could serve as an antagonist. Ideally, the mutation does not reside in a critical binding site for the target molecule but instead at a site that is still vital for function, e.g., an activation region. The inventors have synthesized a series of such mutant peptides substituting alanine for individual amino acid specified in Table 1. Twenty five amino acid substitution mutant peptides built into tat minimal domain peptides or longer peptides were tested for their ability to transactivate HIVLTRCAT by the standard assay procedure. All tat peptides showing deficiency in transactivation, as detailed in Table 3, are potential antagonists. Using this approach, the inventors have identified 14 different HIV tat mutant peptides including several full length tat peptides that exhibit high levels of antagonist activity. These are described in the following sections.
TABLE 3
Transactivation of HIVLTR by Chemically Synthesized HIV Tat Mutant Peptides with Amino Acid Substitutions in Region I or II
Transactivated Cells (%) Total (High Density)
Tat Peptide Added l ϊ lfi' -5 M i x 10 -6 M
Tat 37-62 Mutants 37-62 44 ( 15 ) 28 ( 9 ) 37-62 , 37Ala 40 ( 10 ) 20 ( 3 ) A3
Table 3 (continued)
37-62, 38Ala 27 (4) 10 (0)
37-62, 39Ala 4 (1) 0 (0)
37-62, 40Ala 15 (4) 7 (0)
37-62, 41Ala 5 (0) 0 (0)
37-62,46Ala 0 (0) 0 (0)
37-62, 47Ala 0 (0) 0 (0)
37-62, 46, 47Ala 1 (0) 0 (0)
Tat 37-72 Mutants
37-72 61 (34) 41 (21)
37-72, 39Ala 19 (2) 11 (1)
37-72,40Ala 10 (3) 3 (1)
37-72, 41Ala 2 (0) 0 (0)
37-72, 46Ala 0 (0) 0 (0)
37-72, 47Ala 0 (0) 0 (0)
37-72, 46Ala,47Ala 1 (0) 0 (0)
Tat 37-86 Mutants
37-86 68 (12) 39 (8)
37-86, 41Ala 4 (0) 0 (0)
37-86,47Ala 1 (0) 0 (0)
Tat 1-86 Mutants - chemically synthesized
1-86 72 (56) 54 (23) l-86,22Ala 8 (0) 2 (0) l-86,25Ala 7 (0) 1 (0) l-86,27Ala 2 (1) 0 (0) l-86,30Ala 15 (1) 8 (1) l-86,31Ala 21 (0) 5 (0) l-86,34Ala 55 (15) 49 (8) l-86,37Ala 35 (8) 18 (0) l-86,41Ala 2 (0) 0 (0) l-86,47Ala 7 (0) 1 (0) Table 3 (continued)
Tat 1-86 Mutants - E. coli produced
1-86 59 (20) 33 (10) l-86,22Ser 8 (0) 7 (1)
1-86, 41Ala 8 (2) 4 (0) l-86,46Ala 14 (4) 11 (3) l-86,47Ala 10 (0) 4 (1)
1-86,46, 47Ala 10 (0) 3 (0) l-86,20Thr,46, .47A1;a 10 (3) 5 (0)
The data are the mean of from two to six experiments
1. Strong Tat antagonists are produced by substituting alanin for region II amino acids in peptide backbone tat-dm37-62. Th inventors first substituted alanine for conserved amino acids within the minimal domain peptide tat-dm37-62. In particular, tat peptides with substitutions in amino acids 40 to 47 (regio II, putative activation domain, Figure 6) that are defective i transactivation function (Table 2) were investigated. Tat-dm37 62,41Ala, iai-dm37-62,46Ala, tat-dm37-62.47Ala. and the doubl mutant £&£-dm37-62,Ala46,Ala47 are strong antagonists (Table 4, see Fig. 7). Several of these antagonists reduced wild type tat transactivation activity by 80-90%. Of particular significance, cells expressing large amounts of HIVLTR-directe CAT RNA, i.e., "high density cells," were also reduced by 90%.
TABLE 4
Strong Inhibition by Tat Mutant Peptides of HIV-LTR Transactivation by Tat-86 Peptide Added to Cells
Transactivated Cells Inhibition
Total (High Density)
Tai Peptide 1 S 1QT5 M 1 £ 1__T5 M 5_ x 1Q_" M 2 .1ST* M
Tat : 37-62 Mutants
37- •62, ,37Ala 2 (0) 0 (0) ND ND
10 37- •62, ,39Ala 12 (13) 0 (0) ND ND
37- •62, ,40Ala 47 (39) 4 (4) ND ND
37- ■62, ,41Ala 61 (32) 50 (35) 11 (13) 10 (0)
37- -62, , 46Ala 76 (91) 53 (63) 42 (52) 25 (20) b.
37- •62, , 47Ala 83 (97) 63 (95) 50 (69) 31 (46)
15 37- ■62, ,46,47Ala 85 (89) 67 (50) 41 (33) 15 (0)
The data are the mean of two to twelve experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10""** M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total
20 cell response and 30% high density response. The control values within each experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response.
A
2. Effective tat antagonists are produced by substituting alanine for region II amino acids in peptide backbone tat- dm37-72. Next the same four tat amino acid substitutions wer generated within tat-dm37-72 backbone to determine whether antagonist activity could be increased in this manner. Since the native 37-72 backbone possesses stronger transactivation activity than the native 37-62 backbone, it was considered tha 37-72 may be more effectively taken up by cells or more tightl bound to its target. Consistent with this possibility, all four antagonists were effective, with several showing stronger activity in the 37-72 backbone compared to the same mutant peptides in the 37-62 backbone (see Table 5) .
TABLE 5
Strong Inhibition by Tat Mutant Peptides of HIV-LTR Transactivation by Tat-86 Peptide Added to Cells
Transactivated Cells
%. Inhibition
Total (High Density) τa_t Peptide l ϊ M - 5 M i ___ _ r5 i ___ in-6 M a in - 6 M
Tat 37-72 Mutants
37-72,39Ala 15 (5) 0 (0) ND ND
10 37-72,40Ala 22 (10) 8 (0) ND ND 37-72,41Ala 70 (91) 68 (83) 41 (73) 22 (66) 37-72, 6Ala 76 (100) 62 (91) 47 (74) 37 (60) ? 37-72,47Ala 97 (100) 93 (100) 71 (83) 41 (70) **4 37-72,46,47Ala 82 (100) 54 (33) 21 (44) ND
15 The data are the mean of two to twelve experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10 M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total cell response and 30% high density response. The control values within each
20 experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response.
3. Tat antagonists in peptide backbone tat-dm37-86 containing alanine substituting for region II amino acids are also effective. Two of the substitutions that represent sites for strong antagonist activity, 41Ala and 47Ala, were built into the tat-dm37-86 backbone. Both of these peptides retain good antagonist activity (see Table 6) .
TABLE 6
Strong Inhibition bv Tat Mutant Peptides of HIV-LTR Transactivation by Tat-86 Peptide Added to Cells
. Transactivated Cells
% Inhibition Total (High Density)
Tat Peptide 2. x 10~5 M 1 x lQT5 __M 5_ x 10 M
Tat 37-86 Mutants . 37-86, 1Ala 66 (98) 48 (50) 45 (33)
37-86,47Ala 75 (83) 54 (42) 40 (32)
The data are the mean of two experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10-6 M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total cell response and 30% high density response. The control values within each experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response. A °/
4. Tat antagonists in full length tat-86 backbone containing alanine substituting for region II amino acids. Finally, the same two amino acid substitutions were tested in the full-length tat-86 peptide backbone. Tat-86,41Ala is a strong antagonist (see Table 7). However, Tat-86,47Ala is leaky, i.e. it possesses weak transactivation activity, and accordingly its antagonist activity is diminished, but significant; presumably, the protein conformation imposed by the addition of N-terminal region I alters the activity of region II.
The inventors have tested full-length tat-86 antagonists in a very stringent assay for effectiveness in blocking HIV-LTR-driven gene expression from an integrated HIV-LTRCAT cell line. The results show that in particular tat-86,41Ala is a potent inhibitor of wild-type tat-86 and is apparently stable over the extended period of this assay (48 hrs). Remarkably, £iLt-86,4lAla blocks transactivation 99% at a 4-fold molar excess over that of tat-86. The inventors predict that this particular analog, those listed in Table 1, part 4, and their derivatives will be potent inhibitors of HIV-1 replication.
All of these mutant peptide antagonists are of potential value. Even a somewhat diminished antagonist in the full-length tat backbone may have therapeutic value because of important additional factors including peptide stability and uptake into cells. What is clear from these data is that the same substitutions that produce antagonists in the smallest peptide backbone will likely produce antagonists in larger peptide backbones. Finally, the inventors examined whether substi¬ tutions in other critical amino acids in the full length tat-86 molecule could result in antagonist activity. In principle amino acid substitution in conserved cysteine residues may alter "functional folding" but still permit binding of the mutant peptide to its target. Such molecules could prevent the binding of molecules that possess the active conformation. The full length tat mutant peptides shown in Table 1 are being tested presently for antagonist activity. Several of these exhibit antagonist activity in preliminary studies. Further studies in progress will determine their usefullness as antagonists of HIVLTR trans¬ activation.
TABLE 7
Strong Inhibition by Tat-86 Mutant Peptides of HIV-LTR. Transactivation by Tat-86 Peptide Added to Cells
Transactivated Cells % Inhibition Total (High Density)
Tat Peptide 2 x 1Q~5 M 1 x 10~5 M 5_ x 10~6 M
Tat-86 Mutants - chemically synthesized
Ta_t-86,41Ala 78 (75) 49 (67) 30 (25)
Ta_t-86,47Ala 50 (62) 34 (33) 30 (8)
Tat-86 Mutants - E. coli produced
Ta£-86,41Ala 78 (79) 65 (54) 42 (58)
Tat-86,47Ala 54 (93) 38 (27) 36 (20)
The data are the mean of two experiments. HeLa cells on coverslips were microinjected with pCD12 and treated with Tat-86 (2 x 10~6 M) or Tat-86 together with the indicated concentration of mutant Tat peptide. Treatment with Tat-86 alone gave an average of 54% total cell response and 30% high density response. The control values within each experiment were normalized to 100% for calculation of the percent inhibition of total cell and high density cell response.
To rule out the possibility that inhibition of transactivation by tat antagonists is due in part to non- specific inhibition of transcription, in several experi¬ ments cells on the same coverslip were injected with pSV2CAT or pH4WTCAT which express CAT from the SV40 or human histone 4 promoter, respectively. CAT expression from these pro¬ moters was not affected by tat peptide antagonists. In other experiments, the incorporation of 35S-methionine into an acid-insoluble form was measured in the absence and presence of tat antagonist peptides. No reduction of incorporation was observed at 1 x 10-5 M peptide and less than 10% reduction at 3 x 10-5 M (data not shown) . We conclude that inhibition of HIV-LTR transactivation by tat peptide antagonists is specific and does not reflect a general block in the transcription of injected genes or an inhibition of cellular protein synthesis.
Development of additional tat peptide antagonists. Inasmuch as substitution in tat amino acid residues 41 or 47 generate antagonists of tat-86 in some degree in all peptide backbones tested, the inventors believe and predict that all substitutions in tat-dm37-62 and tat-dm37-72 back¬ bones that have been shown to be antagonists will also be antagonists in longer backbones or in full length tat-86. Furthermore, the inventors believe and predict that inasmuch as 41 and 47 substitutions are generally good antagonists in any backbone, a double substitution of 41 and 47 will be a good antagonist in any backbone. Lastly, the inventors recognize that substitutions with amino acids other than alanine for tat residues 41, 46, 47, 46 and 47, may produce superior antagonists. Gene therapy approach for the delivery of tat antagonist peptides to HIV infected cells. The inventors have synthesized full-length genes encoding some tat peptide antagonists. These mutant tat genes express tat peptides that have been shown to effectively block the function of tat-86. The inventors believe and predict that the expression of these tat mutant genes in cells will result in antagonism of HIV tat function. Such genes therefore are believed to be of value for the gene therapy of AIDS.. The inventors have cloned these tat mutant genes into several viral expression vectors. Transduction into hematopoietic stem cells of AIDS patients is a promising therapy and may avoid problems of drug delivery and drug stability.
ANTAGONISTS OF HUMAN ADENOVIRUS INFECTION Basic Approach. A number of peptides have been synthesized (Table 8 below) for use as potential mutants of th adenovirus ElA oncogene protein domain 3 (PD3). It has been previously demonstrated that the parent peptide, PD3, of 49 amino acids functions as an autonomous transcriptional activato of human adenovirus early genes (Lillie et al., 1987). Further more, it is now known that most of the chemically synthesized mutant peptides are defective in transactivating activity i.7i vivo . These mutant peptides are potential antagonists of PD3 function and may interact with human host cell transcription factors that regulate gene expression. Thus, in the long run the mutant peptides may be useful therapeutically for (1) the inhibition of viral infection in human beings, (2) the regul tion of human gene expression for the possible control of cancer and genetic diseases of man. Furthermore, in vivo and vitro assays have been developed to functionally assay PD3 mutant peptides as possible antagonists of transcriptional activation. Finally, preliminary data suggests that three o the mutant PD3 peptides are antagonists. The application of the method of the present invention to the identification an chemical synthesis of antagonistic peptides to PD3 is describ below.
TABLE 8 POTENTIAL PEPTIDE ANTAGONISTS OF VIRUS INFECTION AND HUMAN GENE EXPRESSION
1. PARENT HUMAN ADENOVIRUS 2 ElA ONCOGENE PROTEIN DOMAIN 3. PD3 (140-188)
NH2-Glu-Glu-Phe-Val-Leu-Asp-Tyr-Val-Glu-His-Pro-Gly-His-Gly-Cys-Arg 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 Ser-Cys-His-Tyr-His-Arg-Arg-Asn-Thr-Gly-Asp-Pro-Asp-Ile-Met-Cys-Ser 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 Leu-Cys-Tyr-Met-Arg-Thr-Cys-Gly-Met-Phe-Val-Tyr-Ser-Pro-Val-Ser-COOH 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188
10 2. AD2 ElA PROTEIN DOMAIN 3 (PD3) SUBSTITUTION MUTANTS (SM)
PD3 parent H. -EEFVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH PD3-154Ala NH. -EEFVLDYVE HPGHGARSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH PD3-157Ala NH2 -EEFVLDYVE HPGHGCRSAH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH PD3-171Alaa NH2 -EEFVLDYVE HYGHGCRSCH YHRRNTGDPD IMASLCYMRT CGMFVYSPVS-COOH
15 PD3-174Ala NH. -EEFVLDYVE HYGHGCRSCH YHRRNTGDPD IMCSLAYMRT CGMFVYSPVS-COOH PD3-174Alaa NH. -EEFVLDYVE HYGHGCRSCH YHRRNTGDPD IMCSLCYMRT AGMFVYSPVS-COOH PD3-174Asn NH. -EEFVLDYVE HYGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYNPVS-COOH
3. AD2 ElA PROTEIN DOMAIN 3 (PD3) N-TERMINAL DELETION MUTANTS (dm)
dml(141-188) NH.-EFVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH
20 dm2(142-288) NH. -FVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH
TABLE 8 (Continued)
dm3(143-188) NH.-VLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH dm4(146-188) NH.-YVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH dm5(148-188) NH.-E HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH 5 dm6(150-188) NH.-PGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH dm7(152-188) NH2-HGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH dm8(156-188) NH.-SCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH dm9(158-188)a NH. -H YHRRNTGDPD IMCSLCYMRT CGMFVYSPVS-COOH
4. AD2 ElA PROTEIN DOMAIN 3 (PD3) C-TERMINAL DELETION MUTANTS (DM)
10 dmlO(140-187) NH2-EEFVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSPV-CONH, dmll(140-186) NH.-EEFVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYSP-CONR, ^ dml2(140-185) NH.-EEFVLDYVE HPGHGCRSCH YHRRNTGDPD IMCSLCYMRT CGMFVYS-CONH,
5. PARENT AD2 ElA PROTEIN DOMAIN 2 (PD2) - TRANSCRIPTIONAL REPRESSION. CELLULAR DNA INDUCTION, AND IMMORTALIZATION DOMAIN
15 PD2-121-138 NH. -DLTCHEAGFPPSDDEDEE-COOH
6. PARENT AD2 ElA PROTEIN DOMAIN 1 (PD1) - TRANSCRIPTION REPRESSION.
CELLULAR DNA INDUCTION, AND IMMORTALIZATION DOMAIN
PD1-41-80 NH.PTLHELYDLDVTAPEDPNEEAVSQIFPDSVMLAVQEGIDL-COOH
Antagonistic activity on ElA transactivation.
30
Step 1 The Development of Sensitive Assays for Biological Activity
To measure the transactivation function of adeno¬ virus ElA PD3 peptides, the inventors have developed two methods of assay which are highly sensitive. First, is an in vivo assay in which a target ElA-inducible gene, early adenovirus gene E2 (present in plasmid pE2) , is co-microin- jected with PD3 peptide or PD3 peptide mutants into HeLa or other suitable cell lines. Transactivation is measured 8 to 18 hours later by immuno-fluorescence which detects the E2 gene product (Lillie et al., 1987) or by in situ hybrid¬ ization which detects viral mRNA. Figure 8 shows the transcriptional activation of the E2 gene by ElA region 3 (Lillie., Loewenstein, Green and Green, 1987) .
Step 2 The Chemical Synthesis of a Large Peptide
Representing the Putative Active Domain
As established by DNA mutational analysis, the ElA 289 ElA protein that functions as a transactivator encodes three functional domains, PD1, PD2, and PD3 (see Figure 9) (Lillie et al., 1986; 1987); only PD3 is essen¬ tial for the transactivation function of the ElA oncogene. Peptides encoding the entire PD3, PD2 and PD1 domains were prepared. It is now known that PD3 alone (49 amino acids, see Table 8) is an autonomous transactivator of early viral genes both in vivo and in vitro (Lillie et al., 1987) .
Step 3 Identification of Smaller Peptides that
Exhibit Transactivating Function by Seguential
Synthesis and Assay of N-Terminal and C-Terminal Deletion Mutants
A series of 9 N-terminal deletion mutants of PD3 were prepared (Table 8, dml to dm9) and tested for in vivo 7 and in vitro transactivation function. All N-terminal deletion mutants exhibited less than full activity by in vivo assay. All activity was lost after deletion of 13 N- terminal amino acids. These mutant peptides are candi- dates for ElA trans- activation antagonists. Three C- terminal deletion mutants have been synthesized (see Table 8).
Step 4 Development of Biological Assays to Measure Peptide Antagonist Function The inventors have shown that an efficient method of introducing PD3 peptides into cells is by co-microinjec¬ tion with the target plasmid encoding an ElA inducible gene. In vivo assays for antagonism involve coinjecting PD3 at a con- centration that exhibits half maximal activity with potential peptide antagonists and the reporter plasmid. Measurement of antagonist activity is performed by assaying the target gene product as described above. In vitro assays for antagonist function involve direct competition between PD3 and potential antagonists using the standard cell-free transcription assay. The level of antagonism is determined by comparison with control in vitro assays using PD3 alone.
Step 5 Identification of Peptide Antagonists among Deletion Mutant Peptides that are Inactive or Possess Low Activity in the Transactivation Assay As mentioned above, several deletion mutant pep¬ tides have been identified that are defective in transacti¬ vation function. These peptides are, therefore, candidate ElA transactivation antagonists. Step 6 Further Development of Peptide Antagonists by Chemical Synthesis of Amino Acid Substitution Mutant Peptides in Minimal Domains
Six single amino acid substitution mutants have been synthesized (see Table 8) . All five cysteines in PD3 have individually been substituted with alanine; four of the cysteines are stringently conserved among different adenovirus serotypes (see Figure 9). All cysteine substi¬ tution mutants were found to be defective in vivo . They fall into two categories, those with substantially reduced activity and two which exhibit no activity at all (PD3- 171Ala and PD3-179Ala, see Table 8). Not unexpectedly, these two cysteine mutants are completely defective in the in vitro assay for transcriptional activation. Preliminary evidence suggest that these two mutant peptides will func¬ tion as antagonists, as determined by the in vitro assay.
ANTAGONISTS OF ONCOGENIC PAPILLOMAVIRUSES
Basic Approach. The method of this invention has been applied to the bovine papillomavirus type 1 E5 gene (Green and Loewenstein, 1987) . This system is important because E5 is an oncogene and is involved in regulating cellular genes involved in cellular DNA replication. A large number of E5, peptides (Table 9 below), some of which represent potential antagonistic peptides of the bovine papillomavirus (BPV1) oncogene E5 have been prepared. It has been demonstrated that the chemically synthesized parent E5 peptide of 44 amino acids and several N-terminal deletion mutant peptides function as autonomous inducers of cellular DNA replication in growth-arrested cells (Green and Loewenstein, 1987) . These peptides may be useful therapeutically for the regulation of human DNA replication, and thus may be of importance for the control of some neo- plastic and genetic diseases of man. The method of the present invention is applied to the bovine papillomavirus E5 protein as described below. 31
Step 1 The Development of Sensitive Assays for Biological Activity
To routinely test the ability of E5 peptide and mutant peptides to induce cellular DNA synthesis, the inventors have used a microinjection assay. Briefly, mouse C127 cells are growth arrested by serum deprivation and then microinjected with E5 peptide preparations using glass capillaries. The induction of DNA synthesis is determined by the incorporation of [3H]thymidine, as measured by auto- radiography (Green and Loewenstein, 1987) .
Step 2 The Chemical Synthesis of a Large Peptide Representing the Putative Active Domain
The entire 44 amino acid E5 protein (see Table 9) was chemically synthesized and demonstrated to function as a potent inducer of cellular DNA synthesis (Green and
Loewenstein, 1987) . About 50% of cells incorporate radio¬ active thymidine after microinjection with as little as 10 ug/ml of the E5 44 amino acid peptide.
Step 3 Identification of Smaller Peptides that Exhibit DNA-Synthesis Inducing Function bv Seouential Synthesis and Assay of N-terminal and C-terminal Deletion Mutants
A series of 14 N-terminal deletion mutants of BPV1 E5 were chemically synthesized (see Table 9) and tested as described above for DNA-synthesis induction function. Remarkably, the C-terminal 13 amino acid peptide was found to be active. The C-terminal 10 and 12 amino acid peptides were devoid of activity. These represent potential E5 antagonists. The sequential addition of 7 amino acids to the C-terminal end substantially increased the DNA induction activity of the resulting peptides. Activity of the C- terminal 20 amino acid peptide (E5-dm 25-44), (see Table 9) was essentially that of the entire 44 amino acid E5 protein. These data demonstrate a minimal core domain of DNA induc¬ tion activity residing in the C-terminal 13 amino acid residues and a near full activity domain residing in the first 20 C-terminal amino acid residues.
Step 4 Development of Biological Assays to Measure Peptide Antagonist Function
The inventors have shown that an efficient method of introducing E5 peptides into growth arrested cells is by microinjection. Assays for antagonism involve co-injecting the E5 44 amino acid peptide at a concentration that exhibits half maximal activity together with potential E5 peptide antagonists. Measurement of antagonist activity is by comparison of DNA induction by the full length E5 44-mer with that of the mixture of E5 44-mer and candidate peptide antagonist.
Step 5 Identification of Peptide Antagonists Among Deletion Mutant Peptides that are Inactive or Possess Low Activity in the Transactivation Assay
As mentioned above, two deletion mutant peptides have been identified that are defective in DNA-synthesis induction function. These peptides are therefore candidate DNA-synthesis induction antagonists.
Step 6 Further Development of Peptide Antagonists by Chemical Synthesis of Amino Acid Substitution Mutant Peptides in E5 Minimal Domains
E5 is conserved among animal papillomavirus type 1 (BPV1) (see Figure 10(a)). Moreover, E5 - s—a-lso conserved among two strong candidates for causative agents of cervical carcinoma and other urogenital malignancies (human papillo- mavirus type 16 (HPV16) and HPV18)r as well as one of the causative agents of venereal condylomas and juvenile laryn- geal papillomatosis (HPV6B) (see Figure 10(b)). <- /
Among the conserved sequences is the hydrophobic stretch which correspond to the essential middle domain of BPVl E5, and the conserved Cys-X-Cys sequence. A number of substitution mutants and N-terminal deletion/ substitution mutants in which Ala is substituted for one or both Cys at residues 37 and 39 have been prepared. Mutants with both substitutions are completely defective in their DNA synthesis-induction function, whereas those with single Cys substitutions are greatly reduced in activity. These substitution mutants may be peptide antagonists of E5 DNA- synthesis induction function.
Human Papillomavirus type 16 E7 oncoprotein. - peptide antagonists
Of considerable medical importance, the E7 oπco- gene of human papillomavirus type 16 (HPV16) encodes a transactivation function (Phelps et al, 1988). The protein encoded by E7 oncogene contains regions of remarkable similarity to those of human adenovirus ElA protein domains 1, 2, and 3 (see Figure 11(a)). The inventors have chemi- cally synthesized the full E7 oncoprotein of 98 amino acids as well as a series of deletion mutant peptides. They have demonstrated that regions 1 and 2 function as an oncogene to induce cellular DNA synthesis (Figure 11(b)). Further¬ more, they have shown that region 3 functions as a trans- activator (Figure 11(c)). Inasmuch as there is much evi¬ dence that HPV16 is involved in cervical cancer and other urogenital malignancies, the inventors predict that peptide antagonists of HPV16 E7 will prove useful for the preven¬ tion and therapy of some human urogenital malignancies. TABLE 9
POTENTIAL PEPTIDE ANTAGONISTS OF CELLULAR DNA REPLICATION
1. PARENT BPVl E5 44-mer
NH. - Met - Pro - Asn - Leu - Trp - Phe - Leu - Leu - Phe - Leu - Gly - 5 ': 1 2 3 4 5 6 7 8 9 10 11
Leu - Val - Ala - Ala - Met - Gin - Leu - Leu - Leu - Leu - Leu - 12 13 14 15 16 17 18 19 20 21 22
Phe - Leu - Leu - Leu - Phe - Phe - Leu - Val - Tyr - Trp - Asp - 23 24 25 26 27 28 29 30 31 32 33
10 His - Phe - Glu - Cys - Ser - Cys - Thr - Gly - Leu - Pro - Phe - COOH. 34 35 36 37 38 39 40 41 42 43 44
2. BPVl E5 PEPTIDE SUBSTITUTION MUTANTS δ
E5(l-44) NH.-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGLPF-COOH
E5-37Ala,39Ala NH.-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FEASATGLPF-COOH
15 E5-43Ala NH.-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGLPF-COOH
E5-34Ala NH.-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDA FECSCTGLPF-COOH
E5-32Ala NH.-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYADH FECSCTGLPF-COOH
3. BPVl E5 PEPTIDE COMBINED N-TERMINAL DELETION (dm) AND SUBSTITUTION MUTANTS
TABLE 9 (Continued)
E5-dm(25-44),43Ala NH.-LLFFLVYWDH FECSCTGLAF-COOH
E5-dm(25-44),34Ala NH,-LLFFLVYWDA FECSCTGLPF-COOH
E5-dm(25-44),32Ala NH.-LLFFLVYADH FECSCTGLPF-COOH
E5-dm(25-44) ,37Ala,39Ala NH,-LLFFLVYWDH FEASATGLPF-COOH
E5-dm(25-44),33Ala NH.-LLFFLVAWDH FECSCTGLPF-COOH
E5-dm(27-44),37Ala NH2-FFLVYWDH FEASCTGLPF-COOH
E5-dm(27-44),37Ala,39Ala NH2-FFLVYWDH FEASATGLPF-COOH
E5-dm(27-44),39Ala, NH.-FFLVYWDH FECSATGLPF-COOH
10 E5-dm(32-44) ,37Ala,39Ala NH.-WDH FEASATGLPF-COOH
E5-dm(32-44),33Ala NH, -WAH FECSCTGLPF-COOH
E5-dm(32-44),34Ala NH.-WDA FECSCTGLPF-COOH δ
BPVl E5 PEPTIDE N-TERMINAL DELETION MUTANTS (dm)
E5-dm(16-44) (29-mer) NH, -MQLLLLLFL LLFFLVYWDH FECSCTGLPF-COOH 15 E5-dm(17-44)(28-mer) NH.-QLLLLLFL LLFFLVYWDH FECSCTGLPF-COOH E5-dm(19-44)(26-mer) NH. -LLLLFL LLFFLVYWDH FECSCTGLPF-COOH E5-dm(21-44) (24-mer) NH,-LLFL LLFFLVYWDH FECSCTGLPF-COOH E5-dm(25-44) (20-mer) NH, -LLFFLVYWDH FECSCTGLPF-COOH E5-dm(27-44) (18-mer) NH. -FFLVYWDH FECSCTGLPF-COOH 20 E5-dm(28-44) (17-mer) NH. -FLVYWDH FECSCTGLPF-COOH E5-dm(29-44) (16-mer) NH. -LVYWDH FECSCTGLPF-COOH
E5-dm(30-44)(15-mer)
Figure imgf000046_0001
E5-dm(31-44)(14-mer) NH,-YWDH FECSCTGLPF-COOH E5-dm(32-44)(13-mer) NH2-WDH FECSCTGLPF-COOH E5-dm(33-44)(12-mer) NH2-DH FECSCTGLPF-COOH E5-dm(35-44)(10-mer) NH, -FECSCTGLPF-COOH
BPVl E5 PEPTIDE C-TERMINAL DELETION MUTANTS (dm)
E5-dm(l-43) NH2-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGLP-CONH, E5-dm(l-42) NH,-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTGL-CONH2 10 E5-dm(l-41) NH2-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCTG-CONH2 E5-dm(l-40) NH2-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSCT-CONH2 E5-dm(1-3,9) NH,-MPNL WFLLFLGLVA AMQLLLLLFL LLFFLVYWDH FECSC-CONH,
BPVl E5 PEPTIDE COMBINED C-TERMINAL AND N-TERMINAL DELETION MUTANTS
E5-dm(25-43) NH. -LLFFLVYWDH FECSCTGLP-COOH 15 E5-dm(25-42) NH2 -LLFFLVYWDH FECSCTGL-COOH E5-dm(25-41) NH. -LLFFLVYWDH FECSCTG-COOH E5-dm(25-40) NH. -LLFFLVYWDH FECSCT-COOH E5-dm(25-39) NH. -LLFFLVYWDH FECSC-COOH
REFERENCES
Arya, S. K., Guo, C, Josephs, S. F. and Wong-Staal, F., 1985. Science 229, 69-73.
Berk, A. J., 1986. Ann. Rev. Genet. 20, 45-79.
Brady, J. N., Bolen, J. B., Radonovich, M. , Salzman, N. and Khoury, G., 1984. Proc. Natl. Acad. Sci. U.S.A. 81, 2040-2044.
Chen, I. S. Y., 1986. Cell 47, 1-2
Chen, I. S. Y., Slamon, D. J., Shah, N. P., Quan, S. G. and Wachsman, W. , 1985. Science 229:54-58.
Clark-Lewis, I., Aebersold, R., Ziltener, H., Schrader, J. W., Hood, L. E. and Kent, S. B. H., 1986. Science 231, 134-139.
Cullen, B. R., 1986, Cell 46, 973.
Dayton, A. I., Sodroski, J. G., Rosen, C. A., Goh, W. C. and Haseltine, W. A., 1986. Cell 44, 941-947.
Derse, D. , 1987. J. Virol. 62, 1115-1119.
Feinberg, M. B., Jarrett, R. F., Aldovini, A., Gallo, R. C. and Wong-Staal, F., 1986. Cell 46, 807.
Felber, B. K. , Paskalis, H., Kleinrnan-Ewing, C, Wong-Staal, F. and Pavlakis, G., 1985. Science 229, 675-679. Fujisawa, J. , Seiki, M. , Sato, M. and Yoshida, M. , 1986. EMBO J. 5, 713-718.
Green, M. and Loewenstein, P. M. , 1987. Cell 51, 795-802.
Hauber, J. , Perkins, A., Heimer, E. P. and Cullen, B. R. , 1987. Proc. Natl. Acad. Sci. U.S.A. 84, 6364.
Hirochika, H., Broker, T. R. and Chow, L. T. , 1987. J. Virol. 61, 2599-2606.
Ihara, S., Feldman, L., Watanabe, S. and Ben-Porat, T.,- 1983. Virology 131, 437-453.
Keller, J. M. and Alwine, J. C, 1984. Cell 36, 381-389.
Kingston, R. E. , Baldwin, A. S. and Sharp, P. A., 1985. Cell 41, 3-5.
Lillie, J. W. r Green, M. and Green, M. R., 1986. Cell 46, 1043-1051.
Lillie, J. W. , Loewenstein, P. M. , Green, M. R. and Green, M. , 1987. Cell 50. 1091-1100.
Maniatis, T. , Goodbourn, S. and Fischer, J. A., 1987. Science 236. 1237-1245.
Merrifield, R. B., 1963. J. Am. Chem. Soc. 85, 2149-2154
Muesing, M. A., Smith, D. H. and Capon, D. J. , 1987. Cell 48, 691-701.
O'Hare, P. and Hayward, G. S., 1985. J. Virol. 61, 190-199. -/I Okamoto, T. and Wong-Staal, F., 1986. Cell 47, 29-35.
Peterlin, B. M., Luciw, P. A., Barr, P. J. and Walker, M. D., 1986. Proc. Natl. Acad. Sci. U.S.A. 83, 9734.
Phelps, W. C. and Howley, P. M. , 1987. J. Virol. 61, 1630-1638.
Phelps, W. C, Yee, C. L. and Howley, P. M. , 1988. Cell 53, 539-547.
Rosen, C, Sodroski, J., Kettmann, R., Burny, A. and Haseltine, W.A. , 1985. Science 227, 320-322.
Rosen, C, Sodroski, J. G., Goh, W. , Dayton, A., Lippke, J. and Haseltine, W. A., 1986. Nature (London) 555-559.
Seiki, M. , Inoue, J., Takeda, T. and Yoshida, M. , 1986. EMBO J. 5, 561-565.
Sodroski, J., Rosen, C, Goh, W. C. and Haseltine, W. , 1985a. Science 228, 1430-1434.
Sodroski, J. G., Rosen, C, Wong-Staal, F., Slahuddin, S. Z., Popovic, M. , Arya, S., Gallo, R. C. and Haseltine, W. A., 1985b. Science 227, 171-173.
Spandau, D. F. and Lee, C. H., 1988. J. Virol. 62, 427-434.
Twu, J. S. and Schloemer, R. H., 1987. J. Virol. 61, 3448-3453.
Wright, C. M. , Felber, B. K. , Paskalis, H. and Pavlakis, G. N., 1986. Science 234, 988.

Claims

WHAT IS CLAIMED IS:
1. A method for preparing antagonists of viral replication which comprises
(a) producing peptide fragments of the transactivating protein of a virus; and (b) selecting said fragment which exhibits antagonist activity toward replication of said virus.
2. A method for preparing antagonists of viral replication which comprises
(a) producing a peptide fragment containing an active domain of the transactivating protein of a virus or a full length peptide of the transactivating protein of a virus; and
(b) making at least one amino acid substitu¬ tion in said peptide fragment containing an active domain or in said full length peptide to deactivate the transactivating activity of the peptide while not destroy¬ ing its binding ability thereby producing an antagonist to replication of said virus.
3. A method of claim 1 in which the virus is selected from the group consisting of. human immuno¬ deficiency virus type 1, herpes simplex virus, human adenovirus, human papillomavirus, and human hepatitis B virus.
4. A method of claim 2 in which the virus is selected from the group consisting of human immuno¬ deficiency virus type 1, herpes simplex virus, human adenovirus, human papillomavirus, and human hepatitis B virus.
5. A method of claim 1 in which the virus is human immunodeficiency virus type 1.
6. A method of claim 2 in which the virus is human immunodeficiency virus type 1.
7. An antagonist produced by the method of claim 5
8. An antagonist produced by the method of claim 6
9. A peptide containing an active domain of the transactivating protein of HIV useful in producing antagonists to HIV selected from the group consisting of tat-86. tat-dm22-86, tat-dm37-86. ±_a_t-dm32-57, tat-37-57. tat-dm37-62 and tat-dm37-72.
10. An antagonist of replication of HIV selected from the group consisting of tat-dm52-86, tat- dm49-57, £a±-dm37-62,41Ala, £a£-dm37-62, 6Ala, tat-dm37- 62,47Ala, £aJt-dm37-62,46Ala,47Ala, tat_-dm37-72,41Ala, Ja£-dm37-72,46Ala, tat-dm37-72, 7Ala, £a±-dm37-72,46Ala, 47Ala, t_a£-dm37-86,41Ala, £a_fc-dm37-86,47Ala, tat-86,41Ala. and tat-86,47Ala.
11. A pharmaceutical composition comprising an antagonist of claim 10 and a pharmaceutical carrier.
PCT/US1989/002404 1988-06-16 1989-06-01 Antagonists of viral transactivating proteins WO1989012461A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US20739388A 1988-06-16 1988-06-16
US207,393 1988-06-16
US35268889A 1989-05-22 1989-05-22
US352,688 1989-05-22

Publications (1)

Publication Number Publication Date
WO1989012461A1 true WO1989012461A1 (en) 1989-12-28

Family

ID=26902211

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1989/002404 WO1989012461A1 (en) 1988-06-16 1989-06-01 Antagonists of viral transactivating proteins

Country Status (2)

Country Link
AU (1) AU3756789A (en)
WO (1) WO1989012461A1 (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992007871A1 (en) * 1990-10-24 1992-05-14 Allelix Biopharmaceuticals Inc. Peptide-based inhibitors of hiv replication
WO1992012728A1 (en) * 1991-01-28 1992-08-06 Biogen, Inc. Repressors of the trans-activating function of papillomavirus e2 proteins
EP0522081A1 (en) * 1990-03-30 1993-01-13 Smithkline Beecham Corporation Inhibition of disease associated with immunodeficiency virus infection
WO1993011230A1 (en) * 1991-12-02 1993-06-10 Dynal As Modified mammalian stem cell blocking viral replication
WO1993021941A1 (en) * 1992-04-23 1993-11-11 Allelix Biopharmaceuticals Inc. Treatment of herpesvirus infection
EP0571549A1 (en) * 1991-02-14 1993-12-01 La Jolla Cancer Research Foundation A NOVEL INTEGRIN SPECIFICITY FOR THE HIV Tat PROTEIN
FR2700169A1 (en) * 1993-01-04 1994-07-08 Transgene Sa New trans-dominant variants of HIV TAT protein
WO1996027389A1 (en) * 1995-03-08 1996-09-12 Neovacs Non-toxic immunogens derived from a retroviral regulatory protein, antibodies, preparation method therefor, and pharmaceutical compositions containing same
FR2731355A1 (en) * 1995-03-08 1996-09-13 Neovacs NOVEL IMMUNOGENS, NEW ANTIBODIES, PREPARATION METHOD AND PHARMACEUTICAL COMPOSITIONS COMPRISING THE SAME
US5633230A (en) * 1990-10-24 1997-05-27 Allelix Biopharmaceuticals, Inc. Treatment of cytomegalovirus infection
US5646120A (en) * 1990-10-24 1997-07-08 Allelix Biopharmaceuticals, Inc. Peptide-based inhibitors of HIV replication
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US5674849A (en) * 1990-10-24 1997-10-07 Allelix Biopharmaceuticals Inc. Anti-viral compositions
WO1998014587A1 (en) * 1996-10-04 1998-04-09 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEAL TH AND HUMAN SERVICES Inhibition of hiv replication using soluble tat peptide analogs
US5804604A (en) * 1989-12-21 1998-09-08 Biogen, Inc. Tat-derived transport polypeptides and fusion proteins
US5831001A (en) * 1990-10-24 1998-11-03 Allelix Biopharmaceuticals Inc. Treatment of herpesvirus infection
WO1999029721A1 (en) * 1997-12-10 1999-06-17 Washington University Anti-pathogen system and methods of use thereof
US6200575B1 (en) 1996-03-07 2001-03-13 Neovacs Non-toxic immunogens derived from a retroviral regulatory protein antibodies preparation process and pharmaceutical compositions comprising them
US6316003B1 (en) 1989-12-21 2001-11-13 Whitehead Institute For Biomedical Research Tat-derived transport polypeptides
US6468969B1 (en) * 1997-08-19 2002-10-22 Tariq M. Rana Inhibition of HIV-1 replication using d-amino acid peptides
US6686333B1 (en) * 1996-10-04 2004-02-03 The United States Of America As Represented By The Department Of Health And Human Resources Inhibition of HIV replication using soluble Tat peptide analogs

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987002989A1 (en) * 1985-11-06 1987-05-21 United States Of America, Represented By The Unite Aids virus gene expression
EP0291893A1 (en) * 1987-05-19 1988-11-23 The Du Pont Merck Pharmaceutical Company Stable human cell lines expressing an indicator gene product under virus-specific genetic controls

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1987002989A1 (en) * 1985-11-06 1987-05-21 United States Of America, Represented By The Unite Aids virus gene expression
EP0291893A1 (en) * 1987-05-19 1988-11-23 The Du Pont Merck Pharmaceutical Company Stable human cell lines expressing an indicator gene product under virus-specific genetic controls

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF VIROLOGY, Vol. 63(3), pp. 1181-87, March 1989, J. HAUBER et al.: "Mutational analysis of the conserved basic domain of human immunodeficiency virus tat protein". *
M.S. GOTTLIEB et al., (editors), "Current Topics in AIDS, Vol. 1, pp. 102-104, John Wiley and Sons, Chichester, UK. Published 1987. *
NATURE, Vol. 320, pp. 367-371, 1986, A.G. FISHER et al.: "The trans-activator gene of HTLV-III is essential for virus replication". See abstract and p. 370. *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, Vol. 85, pp. 6297-6300, 1988, A.D. FRANKEL et al.: "Dimerization of the tat protein from human immunodeficiency virus...". *
PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, Vol. 86, pp. 821-824, 1989, R. GENTZ et al.: "Bioassay for trans-activation using purified human immunodeficiency virus tat-encoded protein...". See p. 823-824 and abstract. *
S. BRODER (editor), "AIDS: Modern Concepts and Therapeutic Challenges", Marcel Dekker Inc., New York and Basel. Published 1987. *
SCIENCE, Vol. 228, pp. 1430-1434, 1985, J.G. SODROSKI et al.: "A transcriptional activator protein encoded by the x-lor region of the human T-cell leukemia virus". See entire article. *
SCIENCE, Vol. 229, pp. 74-77, 1985, J.G. SODROSKI et al.: "Location of the Transactivating region...". *

Cited By (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5747641A (en) * 1989-12-21 1998-05-05 Biogen Inc Tat-derived transport polypeptide conjugates
US5652122A (en) * 1989-12-21 1997-07-29 Frankel; Alan Nucleic acids encoding and methods of making tat-derived transport polypeptides
US6316003B1 (en) 1989-12-21 2001-11-13 Whitehead Institute For Biomedical Research Tat-derived transport polypeptides
US5674980A (en) * 1989-12-21 1997-10-07 Biogen Inc Fusion protein comprising tat-derived transport moiety
US5670617A (en) * 1989-12-21 1997-09-23 Biogen Inc Nucleic acid conjugates of tat-derived transport polypeptides
US5804604A (en) * 1989-12-21 1998-09-08 Biogen, Inc. Tat-derived transport polypeptides and fusion proteins
EP0522081A1 (en) * 1990-03-30 1993-01-13 Smithkline Beecham Corporation Inhibition of disease associated with immunodeficiency virus infection
EP0522081A4 (en) * 1990-03-30 1993-06-16 Smithkline Beecham Corporation Inhibition of disease associated with immunodeficiency virus infection
WO1992007871A1 (en) * 1990-10-24 1992-05-14 Allelix Biopharmaceuticals Inc. Peptide-based inhibitors of hiv replication
US5789531A (en) * 1990-10-24 1998-08-04 Allex Biopharmaceuticals, Inc. Peptide-based inhibitors of HIV replication
US5831001A (en) * 1990-10-24 1998-11-03 Allelix Biopharmaceuticals Inc. Treatment of herpesvirus infection
AU660947B2 (en) * 1990-10-24 1995-07-13 Allelix Biopharmaceuticals Inc. Peptide-based inhibitors of HIV replication
US5674849A (en) * 1990-10-24 1997-10-07 Allelix Biopharmaceuticals Inc. Anti-viral compositions
US5633230A (en) * 1990-10-24 1997-05-27 Allelix Biopharmaceuticals, Inc. Treatment of cytomegalovirus infection
US5646120A (en) * 1990-10-24 1997-07-08 Allelix Biopharmaceuticals, Inc. Peptide-based inhibitors of HIV replication
US5219990A (en) * 1991-01-28 1993-06-15 Biogen, Inc. Papillomavirus e2 trans-activation repressors
US5595884A (en) * 1991-01-28 1997-01-21 Biogen Inc. Papillomavirus E2 transactivation repressor proteins and DNA
US5616559A (en) * 1991-01-28 1997-04-01 Biogen, Inc. Papillomavirus E2 trans-activation repressors
WO1992012728A1 (en) * 1991-01-28 1992-08-06 Biogen, Inc. Repressors of the trans-activating function of papillomavirus e2 proteins
US5656599A (en) * 1991-01-28 1997-08-12 Biogen, Inc. Papillomavirus E2 trans-activation repressors
US5667965A (en) * 1991-01-28 1997-09-16 Biogen, Inc. Papillomavirus E2 trans-activation repressors
EP0571549A4 (en) * 1991-02-14 1995-03-15 Jolla Cancer Res Found A NOVEL INTEGRIN SPECIFICITY FOR THE HIV Tat PROTEIN.
EP0571549A1 (en) * 1991-02-14 1993-12-01 La Jolla Cancer Research Foundation A NOVEL INTEGRIN SPECIFICITY FOR THE HIV Tat PROTEIN
WO1993011230A1 (en) * 1991-12-02 1993-06-10 Dynal As Modified mammalian stem cell blocking viral replication
WO1993021941A1 (en) * 1992-04-23 1993-11-11 Allelix Biopharmaceuticals Inc. Treatment of herpesvirus infection
EP1424394A3 (en) * 1993-01-04 2005-10-05 Transgene S.A. TAT transdominant variants from HIV virus
FR2700169A1 (en) * 1993-01-04 1994-07-08 Transgene Sa New trans-dominant variants of HIV TAT protein
EP0614980A1 (en) * 1993-01-04 1994-09-14 Transgene S.A. TAT transdominant variants from human Immunodeficiency virus
US5889175A (en) * 1993-01-04 1999-03-30 Transgene S.A. Nucleic acids encoding HIV-1 trans-dominant mutants and their use to abrogate HIV-1 viral replication
EP1580272A2 (en) * 1993-01-04 2005-09-28 Transgene S.A. TAT transdominant variants of human immunodeficiency virus (HIV)
EP1580272A3 (en) * 1993-01-04 2005-10-05 Transgene S.A. TAT transdominant variants of human immunodeficiency virus (HIV)
EP1424394A2 (en) * 1993-01-04 2004-06-02 Transgene S.A. TAT transdominant variants from HIV virus
US6132721A (en) * 1995-03-08 2000-10-17 Neovacs Non-Toxic immunogens derived from a retroviral regulatory protein, antibodies, preparation method therefor, and pharmaceutical compositions containing same
WO1996027389A1 (en) * 1995-03-08 1996-09-12 Neovacs Non-toxic immunogens derived from a retroviral regulatory protein, antibodies, preparation method therefor, and pharmaceutical compositions containing same
FR2731355A1 (en) * 1995-03-08 1996-09-13 Neovacs NOVEL IMMUNOGENS, NEW ANTIBODIES, PREPARATION METHOD AND PHARMACEUTICAL COMPOSITIONS COMPRISING THE SAME
US6200575B1 (en) 1996-03-07 2001-03-13 Neovacs Non-toxic immunogens derived from a retroviral regulatory protein antibodies preparation process and pharmaceutical compositions comprising them
WO1998014587A1 (en) * 1996-10-04 1998-04-09 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE SECRETARY, DEPARTEMENT OF HEAL TH AND HUMAN SERVICES Inhibition of hiv replication using soluble tat peptide analogs
US6686333B1 (en) * 1996-10-04 2004-02-03 The United States Of America As Represented By The Department Of Health And Human Resources Inhibition of HIV replication using soluble Tat peptide analogs
US6468969B1 (en) * 1997-08-19 2002-10-22 Tariq M. Rana Inhibition of HIV-1 replication using d-amino acid peptides
US6645501B2 (en) 1997-12-10 2003-11-11 Washington University Anti-pathogen system and methods of use thereof
WO1999029721A1 (en) * 1997-12-10 1999-06-17 Washington University Anti-pathogen system and methods of use thereof

Also Published As

Publication number Publication date
AU3756789A (en) 1990-01-12

Similar Documents

Publication Publication Date Title
WO1989012461A1 (en) Antagonists of viral transactivating proteins
Green et al. Autonomous functional domains of chemically synthesized human immunodeficiency virus tat trans-activator protein
Green et al. Mutational analysis of HIV-1 Tat minimal domain peptides: identification of trans-dominant mutants that suppress HIV-LTR-driven gene expression
AU669843B2 (en) Repressors of the trans-activating function of papillomavirus E2 proteins
Phelps et al. Structure-function analysis of the human papillomavirus type 16 E7 oncoprotein
Ruben et al. Structural and functional characterization of human immunodeficiency virus tat protein
DiMaio et al. The E5 proteins
Cesarman et al. Kaposi's sarcoma-associated herpesvirus contains G protein-coupled receptor and cyclin D homologs which are expressed in Kaposi's sarcoma and malignant lymphoma
Garcia-Carranca et al. Interplay of viral and cellular proteins along the long control region of human papillomavirus type 18
Dalal et al. Mutational analysis of human papillomavirus type 16 E6 demonstrates that p53 degradation is necessary for immortalization of mammary epithelial cells
CA1326459C (en) Diagnostic peptides of human papilloma virus
Conrad et al. The human papillomavirus type 6 and 16 E5 proteins are membrane-associated proteins which associate with the 16-kilodalton pore-forming protein
JP3859696B2 (en) DNA sequence derived from the genome of papillomavirus HPV39, application of said sequence to the generation of IN VITRO diagnosis and immunogenic composition
Desai et al. Isolation of a cellular protein that binds to the human immunodeficiency virus Tat protein and can potentiate transactivation of the viral promoter.
Rawls et al. Chemical synthesis of human papillomavirus type 16 E7 oncoprotein: autonomous protein domains for induction of cellular DNA synthesis and for trans activation
AU668441B2 (en) New transdominant TAT variants of the human immunodeficiency virus
Green et al. Demonstration that a chemically synthesized BPV1 oncoprotein and its C-terminal domain function to induce cellular DNA synthesis
Taylor et al. E1A-mediated inhibition of myogenesis correlates with a direct physical interaction of E1A12S and basic helix-loop-helix proteins
Yin et al. Chimeric proteins composed of Jun and CREB define domains required for interaction with the human T-cell leukemia virus type 1 Tax protein
CA2077322A1 (en) Peptide inhibitors of human papilloma virus protein binding to retinoblastoma gene proteins
Dixon et al. The E1 helicase of human papillomavirus type 11 binds to the origin of replication with low sequence specificity
WO1994015634A1 (en) Tat and rev oligopeptides in hiv treatment
WO1994015634A9 (en) Tat and rev oligopeptides in hiv treatment
US6673354B2 (en) Compositions and methods for treating papillomavirus-infected cells
Watanabe et al. The E7 functions of human papillomaviruses in rat 3Y1 cells

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE BF BJ CF CG CH CM DE FR GA GB IT LU ML MR NL SE SN TD TG