EP0644946A1 - Vector particles resistant to inactivation by human serum - Google Patents

Vector particles resistant to inactivation by human serum

Info

Publication number
EP0644946A1
EP0644946A1 EP93913964A EP93913964A EP0644946A1 EP 0644946 A1 EP0644946 A1 EP 0644946A1 EP 93913964 A EP93913964 A EP 93913964A EP 93913964 A EP93913964 A EP 93913964A EP 0644946 A1 EP0644946 A1 EP 0644946A1
Authority
EP
European Patent Office
Prior art keywords
protein
amino acid
changed
vector particle
mutated
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP93913964A
Other languages
German (de)
French (fr)
Other versions
EP0644946A4 (en
Inventor
W. French Anderson
James M. Mason
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Original Assignee
US Department of Health and Human Services
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 US Department of Health and Human Services filed Critical US Department of Health and Human Services
Publication of EP0644946A1 publication Critical patent/EP0644946A1/en
Publication of EP0644946A4 publication Critical patent/EP0644946A4/en
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12N2740/00Reverse transcribing RNA viruses
    • C12N2740/00011Details
    • C12N2740/10011Retroviridae
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13022New 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
    • C12N2740/13011Gammaretrovirus, e.g. murine leukeamia virus
    • C12N2740/13041Use of virus, viral particle or viral elements as a vector
    • C12N2740/13043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vector

Definitions

  • This invention relates to "injectable" vect particles. More particularly, this invention relates vector particles, such as retroviral vector particle wherein such vector particles are resistant to inactivati by human serum.
  • Vector particles are useful agents for introduci gene(s) or DNA (RNA) into a cell, such as a eukaryotic cel
  • the gene(s) is controlled by an appropriate promote
  • vectors which may be employed to generate vect particles include prokaryotic vectors, such as bacteri vectors; eukaryotic vectors, including fungal vectors su as yeast vectors; and viral vectors such as DNA vir vectors, RNA virus vectors, and retroviral vector Retroviruses which have been employed for generating vect particles for introducing genes or DNA (RNA) into a ce include Moloney Murine Leukemia Virus, Spleen Necros Virus, Rous Sarcoma Virus and Harvey Sarcoma Virus.
  • T term "introducing” as used herein encompasses a variety methods of transfering genes or DNA (RNA) into a cell. Su methods include transformation, transduction, transfectio and infection.
  • Vector particles have been used for introducing D (RNA) into cells for gene therapy purposes.
  • RNA D
  • Such a procedure involves obtaining cells from a patient a using a vector particle to introduce desired DNA (RNA) i the cells and then providing the patient with the engineer cells for a therapeutic purpose.
  • RNA DNA
  • Such alternative procedure would involve genetically engineeri cells jLn vivo.
  • a vector particle whic includes the desired DNA (RNA) would be administered to patient for .in vivo delivery to the cells of a patient.
  • an object of the present invention t provide gene therapy by introduction of a vector particle such as, for example, a retroviral vector particle, into patient, wherein the vector particle is resistant t inactivation by human serum.
  • a vector particle such as, for example, a retroviral vector particle
  • the vecto particle is a viral vector particle, and more preferably th viral vector particle is a retroviral vector particle.
  • retroviruses include a protei known as pl5E, and Applicants have found that retroviruse are susceptible to inactivation by human serum as a resul of the action of complement protein(s) present in serum o the pl5E protein portion of the retroviru ⁇ . Applicants hav further found that such retroviruses can be made resistan to inactivation by human serum by mutating such pl5 protein.
  • a retrovira vector wherein a portion of the DNA encoding pl5E protei (shown in the accompanying sequence listing), has bee mutated to render the vector particle resistant t inactivation by human serum.
  • the terms "mutated” an “mutation” as used herein mean that the DNA encoding pl5 protein has been changed such that at least one but not al of the amino acids of pl5E protein have been changed (suc changes can include point mutations, deletions, and/o insertions) .
  • pl5E protein is a viral protein having 196 amino aci residues.
  • viruses can contain both the pl5E and pl2 proteins.
  • pl5E protein is anchored in the viral membran such that amino acid residues 1 to 134 are present on th outside of the virus.
  • the pl5E protein includes tw regions, amino acid residues 39 to 61 (sometimes hereinafte referred to as region 1), and amino acid residues 101 to 12 (sometimes hereinafter referred to as region 2), whic Applicants believe have an external location in th three-dimensional structure of the pl5E protein; i.e., suc regions are directly exposed to human serum.
  • Region 2 is highly conserved region in many retroviruses, even thoug the amino acid sequences of this region are not identical i all retroviruses.
  • Such regions are complement bindin regions. Examples of complement proteins which may bind t the complement binding regions are CIS and C1Q, which bin to regions 1 and 2.
  • complement protein bind to both region 1 and region 2.
  • at least one portion of DNA encoding complement binding region of pl5E protein has been mutated Such a mutation results in a change of at least one amin acid residue of a complement binding region of pl5E protein
  • the change in at least one amino acid residue of complement binding region of pl5E protein prevents bindin of a complement protein to the complement binding region thereby preventing complement inactivation of th retrovirus.
  • at least one amino aci residue in both complement binding regions of pl5E prote is changed, whereas in another embodiment, at least o amino acid residue in one of the complement binding regio is changed.
  • the at least one portion of D encoding pl5E protein is mutated such that at least o positively charged amino acid residue or negatively charg amino acid residue is changed to an amino acid resid having the opposite charge.
  • the positively charged amino acids are His, Lys, a Arg.
  • the negatively charged amino acids are Asp and Glu.
  • the at least one portion of D encoding pl5E protein is mutated such that at least o positively charged amino acid or negatively charged ami acid is changed to a noncharged amino acid.
  • the at least one portion of D encoding a complement binding region of pl5E protein whi is mutated, encodes one or more of amino acid residues 1 to 123 of pl5E protein.
  • the at least o portion of DNA encoding pl5E protein is mutated such th amino acid residue 122 is changed.
  • the at least one portion of D encoding pl5E protein is mutated such that at least one amino acid residues 117 and 122 are changed.
  • Preferabl amino acid residue 117 is changed from Arg to Glu
  • ami acid residue 122 is changed from Glu to Gin.
  • the at least one portion of D encoding pl5E protein ⁇ Yts mutated such that amino ac residues 104, 105, 109, and 111 are changed.
  • Preferabl amino acid residue 104 is changed from Arg to His
  • ami acid residue 105 is changed from Asp to Asn
  • amino ac residue 109 is changed from Lys to Gin
  • amino ac residue ill is changed from Arg to Gin.
  • the at least one portion of D encoding pl5E protein is mutated such that amino ac residues 104, 105, 109, 111, 117, and 122 are change
  • the at least one portion of DNA is mutated su that amino acid residue 104 is changed from Arg to Hi amino acid residue 105 is changed from Asp to Asn, ami acid residue 109 is changed from Lys to Gin, amino ac residue 111 is changed from Arg to Gin, amino acid resid 117 is changed from Arg to Glu, and amino acid residue 1 is changed from Glu to Gin.
  • the mutation DNA encoding pl5E protein may be effected by deleting portion of the pl5E gene, and replacing the deleted porti of the pl5E gene, with fragment(s) or portion(s) of a ge encoding another viral protein.
  • o portion of DNA encoding the pl5E protein is replaced with fragment of the gene encoding the p21 protein, which is HTLV-I transmembrane protein.
  • HTLV-I virus has been fou to be resistant to binding by complement proteins and th HTLV-I is resistant to inactivation by human serum (Hoshin et al., Nature, Vol. 310, pgs. 324-325 (1984)).
  • a retroviral vect particle wherein a portion of the pl5E protein has be deleted and replaced with a portion of another vir protein, such as a portion of the p21 protein.
  • p21 protein (as shown in the accompanying sequenc listing) is a protein having 176 amino acid residues, an which, in relation to pl5E, has significant amino aci sequence homology.
  • at least amino aci residues 39 to 61, and 101 to 123 are deleted from pl5 protein, and replaced with amino acid residues 34 to 56 an 96 to 118 of p21 protein.
  • at leas amino acid residues 39 to 123 of pl5E protein are delete and replaced with amino acid residues 34 to 118 of p2 protein.
  • amino acid residues 39 to 69 o pl5E protein are deleted and replaced with amino aci residues 34 to 64 of p21 protein, and amino acid residues 9 to 123 of pl5E protein are deleted and replaced with amin acid residues 91 to 118 of p21 protein.
  • Vector particles thus generated, and which ar resistant to inactivation by human serum, may be engineere such that the vector particles may, when introduced into patient, travel directly to a target cell or tissue.
  • the vector particle furthe includes a protein which contains a receptor binding regio that binds to a receptor of a human target cell, such as for example, but not limited to, the amphotropic cel receptor.
  • retroviral vectors hereinabove described may b constructed by genetic engineering techniques known to thos skilled in the art.
  • the retroviral vector may be of th LN series of vectors, as described in Bender, et al. J.Virol., Vol. 61, pgs. 1639-1649 (1987) and Miller, et al. Biotechniques, Vol. 7, pgs. 980-990 (1989).
  • the retroviral vector includes multiple restriction enzyme site, or multiple cloning site
  • the multiple cloning site includes at least four cloning, o restriction enzyme sites, wherein at least two of the site have an average frequency of appearance in eukaryotic gene of less than once in 10,000 base pairs; i.e., th restriction product has an average size of at least 10,00 base pairs.
  • restriction sites also sometime hereinafter referred to as "rare" sites, which have a average frequency of appearance in eukaryotic genes of les than once in 10,000 base pairs, contain a CG doublet withi their recognition sequence, such doublet appearin particularly infrequently in the mammalian genome.
  • Anothe measure of rarity or scarcity of a restriction enzyme sit in mammals is its representation in mammalian viruses, suc as SV40.
  • an enzyme whose recognition sequenc is absent in SV40 may be a candidate for being a "rare mammalian cutter.
  • restriction enzyme sites having an averag frequency of appearance in eukaryotic genes of less tha once in 10,000 base pairs include, but are not limited t the NotI, SnaBI, Sail, Xhol, Clal, Sad, EagI, and Sma sites.
  • Preferred cloning sites are selected from the grou consisting of NotI, SnaBI, Sail, and Xhol.
  • the multiple cloning site has a length n greater than about 70 base pairs, and preferably no greate than about 60 base pairs.
  • the multipl restriction enzyme site, or multiple cloning site is locate between the 5'LTR and 3' TR of the retroviral vector.
  • Th 5' end of the multiple cloning site is no greater than abou 895 base pairs from the 3' end of the 5' LTR, and preferabl at least about 375 base pairs from the 3' end of the 5' LTR
  • the 3* end of the multiple cloning site is no greater tha about 40 base pairs fro... the 5 1 end of the 3' LTR, an preferably at least 11 base pairs from the 5' end of the 3 LTR.
  • Such vectors may be engineered from existing retrovira vectors through genetic engineering techniques known in th art such that the resulting retroviral vector includes a least four cloning sites wherein at least two of the clonin sites are selected from the group consisting of the NotI SnaBI, Sail, and Xhol cloning sites.
  • the retroviral vector includes each of the NotI SnaBI, Sail, and Xhol cloning sites.
  • Such a retroviral vector may serve as part of a clonin system for the transfer of genes to eukaryotic cells.
  • a cloning system for the manipulatio of genes in a retroviral vector which includes a retrovira vector including a multiple cloning site of the typ hereinabove described, and a shuttle cloning vector whic includes at least two cloning sites which are compatibl with at least two cloning sites selected from the grou consisting of NotI, SnaBI, Sail, and Xhol located on th retroviral vector.
  • the shuttle cloning vector also include at least one desired gene which is capable of bein transferred from said shuttle cloning vector to sai retroviral vector.
  • the shuttle cloning vector may be constructed from basic "backbone" vector or fragment to which are ligated on or more linkers which include cloning or restriction enzym recognition sites. Included in the cloning sites are th compatible, or complementary cloning sites hereinabov described. Genes and/or promoters having ends correspondin to the restriction sites of the shuttle vector may b ligated into the shuttle vector through techniques known i the art.
  • the shuttle cloning vector can be employed to amplif DNA sequences in prokaryotic systems.
  • the shuttle cloni vector may be prepared from plasmids generally used i prokaryotic systems and in particular in bacteria. Thus for example, the shuttle cloning vector may be derived fro plasmids such as pBR322; pUCl ⁇ ; etc.
  • the DNA encoding pl5E protein whic has been mutated to render a vector particle resistant t inactivation by human serum may be contained in a expression vehicle other than a retroviral vector.
  • Suc expression vehicles include, for example, viral vecto other than retroviral vectors, or any expression plasmi which is capable of being transferred into a cell line whic is capable of producing vector particles which include t mutated pl5E protein.
  • Such vectors or expression vehicles which contain D encoding a mutated env protein such as the mutated pl5 protein hereinabove described, are transferred into pre-packaging cell line to generate vector particles.
  • the pre-packaging cell line contains the gag an pol proteins of the virus, plus a retroviral vector lackin the structural gag, pol, and env proteins.
  • An example o such a pre-packaging cell line is the GPL pre-packaging cel line which consists of an NIH 3T3 mouse fibroblast cell li which contains an expression plasmid for MoMuLV gag-po protein as well as the retroviral vector LNL6 (Miller, al., Biotechniques, Vol. 7, pgs. 980-990 (1989)). It is be understood, however, that the scope of the prese invention is not to be limited to any particul pre-packaging cell line.
  • the pre-packaging cell line Upon transfection of the pre-packaging cell line wi an expression vehicle containing DNA encoding a mutated e protein, the pre-packaging cell line will generate vector particles. The vector particles are then tested for complement resistance. The vector particles which are shown to be complement resistant (i.e., not inactivated by human serum), therefore, contain complement resistant envelope proteins encoded by a specific envelope expression vehicle.
  • Such an expression vehicle can then be used, by techniques known to those skilled in the art, to produce a packaging cell line which contains an expression vehicle encoding the retroviral gag and pol proteins, and an expression vehicle containing a gene encoding the mutated env protein (such as, for example, an expression vehicle or expression plasmid containing a mutated pl5E protein such as hereinabove described), whereby such packaging cell line may be employed to generate vector particles which are resistant to inactivation by human serum.
  • a retroviral vector which lacks the structural gag, pol, and env genes, but includes a desired gene of interest, may be transferred into such a packaging cell line.
  • the packaging cell line may generate vector particles which contain a desired gene(s) of interest, and which are resistant to inactivation by human serum.
  • the vector particles generated from the packaging cell line will not be inactivated when contacted with human serum; and in addition, such vector particles, when engineered with protein containing a receptor binding region for a human receptor, are targetable, whereby the receptor binding region for a human receptor enables the vector particles to bind to a target cell.
  • retroviral vector particles may be directly introduced into the body (e.g., by intravenous, intramuscular, or subcutaneous injection, intranasally, orally, rectally or vaginally), and travel to a desired target cell.
  • Such vector particles therefore, are useful for the introduction of desire heterologous genes into target cells in vivo as a gen therapy procedure.
  • the vectors of the present inventio further include at least one heterologous gene
  • Heterologous or foreign genes which may be placed into th vector or vector particles include, but are not limited to genes which encode cytokines or cellular growth factors such as lymphokines, which are growth factors fo lymphocytes.
  • Other examples of foreign genes include, bu are not limited to, genes encoding Factor VIII, Factor IX tumor necrosis factors (TNF's), ADA, ApoE, ApoC, and Protei C.
  • the vectors of the present invention include one o more promoters.
  • Suitable promoters which may be employe include, but are not limited to, the retroviral LTR; th SV40 promoter; and the human cytomegalovirus (CMV) promote described in Miller, et al., Biotechniques, Vol. 7, No. 9 pgs. 980-990 (1989), or any other promoter (eg., cellula promoters such as eukaryotic cellular promoters including but not limited to, the histone, pol III, and ⁇ -acti promoters).
  • Other viral promoters which may be employe include, but are not limited to adenovirus promoters, T promoters, and B19 parvovirus promoters. The selection of suitable promoter will be apparent to those skilled in th art from the teachings contained herein.
  • the vectors of the present invention may contai regulatory elements, where necessary, to ensure tissu specific expression of the desired heterologous gene(s) and/or to regulate expression of the heterologous gene(s) i response to cellular or metabolic signals.
  • retroviral vector particles other viral vector particle (such as, for example, adenovirus and adeno-associated vir particles), or synthetic particles may be construct wherein a region of the envelope protein in the vect particle may be mutated such that the vector partic becomes resistant to inactivation by human serum, there making such vector particles suitable for in vi administration.
  • viral vector particle such as, for example, adenovirus and adeno-associated vir particles
  • synthetic particles may be construct wherein a region of the envelope protein in the vect particle may be mutated such that the vector partic becomes resistant to inactivation by human serum, there making such vector particles suitable for in vi administration.
  • Plasmid pCE2 was constructed from pBR322 such that t resulting plasmid pCE2 includes genes encoding the envelo proteins gp70 and pl5E.
  • pBR322 ( Figure 1) was cut wi EcoRI and filled in to destroy the EcoRI site to gi pBR322Z Rl.
  • pBR3224Rl was then cut with Ndel and filled to destroy the Ndel site to give pBR322
  • a R pBR322_4 R N was digested with Hindi11 and EcoRV, a cloned into the Hindlll/EcoRV fragment was a Hindlll/Fs cassette containing the gp70 and pl5E genes under t control of a cytomegalovirus (CMV) intermediate ear promoter with a polyA (adenine) tail from SV40 ( Figure from plasmid pCEE. ( Figure 3).
  • CMV cytomegalovirus
  • the Hindlll/Fspl casset obtained from plasmid pCEE contains a CMV intermediate ear promoter in which the Ball/Sac11 (bp 21 to bp 766) w converted to an Hindlll/Sall fragment by linker additio the ecotropic envelope Bglll/Nhel fragment (bp 5408 to 7847 of MoMuLV, encoding gp70 and pl5E) was filled and Eco linkers were added; and the SV40 poly A signal from Bell BamHI (bp 2770 to bp 2533) was cloned into a BamHI si (thereby destroying the Bell site). A Bglll site was add at the 3 1 end of the gp70 gene. (This addition does n alter any amino acids).
  • the resulting plasmid is pCE ( Figure 4. )
  • pUC-E2 sub-loning i carried out in a different plasmid called pUC-E2 ( Figure 6.)
  • This plasmid is pUC18 ( Figure 5) with the PvuI fragment removed and replaced with EcoRI linkers.
  • Int the EcoRI site was cloned the MoMuLV ecotropic envelope gen (i.e., the gp70 and pl5E genes from pCE2) from the Bgll site (5408) to the Nhel site (7847), which have been blunte and had EcoRI linkers added.
  • the resulting pUC-E2 plasmi ( Figure 6) therefore has unique Bglll, Spel, Clal, and PvuI sites in and around the pl5E gene.
  • PCR primers are then synthesized to encode th following mrtations in the pl5E protein (using pCE2 as template):
  • Amino acid residue 117 is changed from Arg to Glu and amino acid residue 122 is changed from Glu to Gin;
  • Amino acid residue 104 is changed from Arg to His amino acid residue 105 is changed from Asp to Asn, amin acid residue 109 is changed from Lys to Gin, and amino aci residue 111 is changed from Arg to Gin;
  • Amino acid residue 104 is changed from Arg to His amino acid residue 105 is changed from Asp to Asn, amin acid residue 109 is changed from Lys to Gin, amino aci residue 111 is hanged from Arg to Gin, amino acid residu 117 is changed from Arg to Glu, and amino acid residue 12 is changed from Glu to Gin.
  • Each PCR product is digested with Spel and PvuII, a cloned into pUC-E2 at the unique Spel and PvuII sites. T resulting plasmids are then sequenced to confirm the poi mutations. DNA fragments bearing these mutations are the subcloned into the expression plasmid pCE2.
  • pCE2 i digested with EcoRI and the envelope DNA fragment is remove and replaced with the EcoRI envelope fragment from th pUC-E2 plasmids.
  • the resulting pCE2 plasmids are the checked for orientation of the EcoRI fragment and sequence again (only at the cloning site junctions and at the region bearing the point mutations) to confirm the presence of th newly created mutated pl5E genes.
  • the resulting expressio plasmids are identified as follows: pCR68 - includes mutations in which amino acid residu 117 is changed from Arg to Glu, and amino acid residu 122 is changed from Glu to Gin; pCR69 - includes mutations in which amino acid residu 104 is changed from Arg to His, amino acid residue 10 is changed from Asp to Asn, amino acid residue 109 i changed from Lys to Gin, and amino acid residue 111 i changed from Arg to Gin; and pCR70 - includes mutations in which amino acid residu 104 is changed from Arg to His, amino acid residue 10 is changed from Asp to Asn, amino acid residue 109 i changed from Lys to Gin,
  • Plasmids pCR68, pCR69, and pCR70 are transfecte separately into the GPL pre-packaging cell line.
  • the GP pre-packaging cell line consists of an NIH 3T3 mous fibroblast cell line which contains an expression plasmi for MoMuLV gag-pol protein as well as the retroviral vecto LNL6 (Miller, et al., 1989).
  • the GPL packaging cell line produces vecto particles. Transiently expressed vector particles are collecte with cell supernatant at 48-72 hrs. post-transfection.
  • the vector particles generated as hereinabove describ may then be assayed for vector titer by techniques known those skilled in the art.
  • the vector particles may also collected in viral supernatant and concentrated, i necessary, according to procedures known to those skilled i the art in order to employ such vector particles in assa or in therapeutic procedures.
  • Advantages of the present invention include the abili to introduce vector particles directly into a human patie whereby the vector particle is not lysed or inactivated human serum upon such introduction.
  • the vect particles of the present invention enable one to deliv desired genes to a patient in vivo.
  • Such vector particl may also be engineered such that they are "targetable", well as injectable, thereby enabling the vector particles travel directly to a target cell or tissue without bei lysed or inactivated by human serum.
  • ADDRESSEE Carella, Byrne, Bain,
  • Glu Lys Ser lie Ser Asn Leu Glu Lys Ser
  • Lys Asp Arg lie Ser Val Val Gin Ala Leu

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Virology (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Microbiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Medicinal Chemistry (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

A retroviral vector particle resistant to inactivation by human serum. The vector particles preferably include p15E protein wherein at least a portion of the DNA encoding p15E protein is mutated such that the vector particle is resistant to inactivation by human serum. The vector particles may further include a protein containing a receptor binding region which binds to the receptor of a human target cell, thereby enabling the direct introduction of desired heterologous genes in vivo, whereby the vector particle including the heterologous gene travels directly to a targeted cell or tissue.

Description

VECTOR PARTICLES RESISTANT TO INACTIVATION BY HUMAN SERUM
This invention relates to "injectable" vect particles. More particularly, this invention relates vector particles, such as retroviral vector particle wherein such vector particles are resistant to inactivati by human serum.
Vector particles are useful agents for introduci gene(s) or DNA (RNA) into a cell, such as a eukaryotic cel The gene(s) is controlled by an appropriate promote Examples of vectors which may be employed to generate vect particles include prokaryotic vectors, such as bacteri vectors; eukaryotic vectors, including fungal vectors su as yeast vectors; and viral vectors such as DNA vir vectors, RNA virus vectors, and retroviral vector Retroviruses which have been employed for generating vect particles for introducing genes or DNA (RNA) into a ce include Moloney Murine Leukemia Virus, Spleen Necros Virus, Rous Sarcoma Virus and Harvey Sarcoma Virus. T term "introducing" as used herein encompasses a variety methods of transfering genes or DNA (RNA) into a cell. Su methods include transformation, transduction, transfectio and infection.
Vector particles have been used for introducing D (RNA) into cells for gene therapy purposes. In genera such a procedure involves obtaining cells from a patient a using a vector particle to introduce desired DNA (RNA) i the cells and then providing the patient with the engineer cells for a therapeutic purpose. It would be desirable provide alternative procedures for gene therapy. Such alternative procedure would involve genetically engineeri cells jLn vivo. In such a procedure, a vector particle whic includes the desired DNA (RNA) would be administered to patient for .in vivo delivery to the cells of a patient.
It is therefore an object of the present invention t provide gene therapy by introduction of a vector particle such as, for example, a retroviral vector particle, into patient, wherein the vector particle is resistant t inactivation by human serum.
In accordance with an aspect of the present invention there is provided a vector particle which is resistant t inactivation by human serum. Preferably, the vecto particle is a viral vector particle, and more preferably th viral vector particle is a retroviral vector particle.
The envelope portion of retroviruses include a protei known as pl5E, and Applicants have found that retroviruse are susceptible to inactivation by human serum as a resul of the action of complement protein(s) present in serum o the pl5E protein portion of the retroviruε. Applicants hav further found that such retroviruses can be made resistan to inactivation by human serum by mutating such pl5 protein.
In one embodiment, there is provided a retrovira vector wherein a portion of the DNA encoding pl5E protei (shown in the accompanying sequence listing), has bee mutated to render the vector particle resistant t inactivation by human serum. The terms "mutated" an "mutation" as used herein mean that the DNA encoding pl5 protein has been changed such that at least one but not al of the amino acids of pl5E protein have been changed (suc changes can include point mutations, deletions, and/o insertions) . pl5E protein is a viral protein having 196 amino aci residues. In viruses, sometimes all 196 amino acid residue are present, and other times, amino acid residues 181 to 19 (known as the "r" peptide), are not present, and th resulting protein is the "mature" form of pl5E known a pl2E. Thus, viruses can contain both the pl5E and pl2 proteins. pl5E protein is anchored in the viral membran such that amino acid residues 1 to 134 are present on th outside of the virus. Although the present invention is no to be limited to any of the following reasoning, Applicant believe complement proteins may bind to this region whereb such binding leads to inactivation and/or lysis of th retrovirus. In particular, the pl5E protein includes tw regions, amino acid residues 39 to 61 (sometimes hereinafte referred to as region 1), and amino acid residues 101 to 12 (sometimes hereinafter referred to as region 2), whic Applicants believe have an external location in th three-dimensional structure of the pl5E protein; i.e., suc regions are directly exposed to human serum. Region 2 is highly conserved region in many retroviruses, even thoug the amino acid sequences of this region are not identical i all retroviruses. Such regions are complement bindin regions. Examples of complement proteins which may bind t the complement binding regions are CIS and C1Q, which bin to regions 1 and 2.
In order to inactivate the retrovirus, complement protein bind to both region 1 and region 2. Thus, in a preferre embodiment, at least one portion of DNA encoding complement binding region of pl5E protein has been mutated Such a mutation results in a change of at least one amin acid residue of a complement binding region of pl5E protein The change in at least one amino acid residue of complement binding region of pl5E protein prevents bindin of a complement protein to the complement binding region thereby preventing complement inactivation of th retrovirus. In one embodiment, at least one amino aci residue in both complement binding regions of pl5E prote is changed, whereas in another embodiment, at least o amino acid residue in one of the complement binding regio is changed.
It is to be understood, however, that the entire D sequence encoding pl5E protein cannot be mutated becau such a change renders the vectors unsuitable for in vi use.
In one embodiment, the at least one portion of D encoding pl5E protein is mutated such that at least o positively charged amino acid residue or negatively charg amino acid residue is changed to an amino acid resid having the opposite charge.
The positively charged amino acids are His, Lys, a Arg.
The negatively charged amino acids are Asp and Glu.
In another embodiment, the at least one portion of D encoding pl5E protein is mutated such that at least o positively charged amino acid or negatively charged ami acid is changed to a noncharged amino acid.
In one embodiment, the at least one portion of D encoding a complement binding region of pl5E protein, whi is mutated, encodes one or more of amino acid residues 1 to 123 of pl5E protein. In one embodiment, the at least o portion of DNA encoding pl5E protein is mutated such th amino acid residue 122 is changed.
In one embodiment, the at least one portion of D encoding pl5E protein is mutated such that at least one amino acid residues 117 and 122 are changed. Preferabl amino acid residue 117 is changed from Arg to Glu, and ami acid residue 122 is changed from Glu to Gin. In another embodiment, the at least one portion of D encoding pl5E protein~Yts mutated such that amino ac residues 104, 105, 109, and 111 are changed. Preferabl amino acid residue 104 is changed from Arg to His, ami acid residue 105 is changed from Asp to Asn, amino ac residue 109 is changed from Lys to Gin, and amino ac residue ill is changed from Arg to Gin.
In another embodiment, the at least one portion of D encoding pl5E protein is mutated such that amino ac residues 104, 105, 109, 111, 117, and 122 are change Preferably, the at least one portion of DNA is mutated su that amino acid residue 104 is changed from Arg to Hi amino acid residue 105 is changed from Asp to Asn, ami acid residue 109 is changed from Lys to Gin, amino ac residue 111 is changed from Arg to Gin, amino acid resid 117 is changed from Arg to Glu, and amino acid residue 1 is changed from Glu to Gin.
In yet another alternative embodiment, the mutation DNA encoding pl5E protein may be effected by deleting portion of the pl5E gene, and replacing the deleted porti of the pl5E gene, with fragment(s) or portion(s) of a ge encoding another viral protein. In one embodiment, o portion of DNA encoding the pl5E protein is replaced with fragment of the gene encoding the p21 protein, which is HTLV-I transmembrane protein. HTLV-I virus has been fou to be resistant to binding by complement proteins and th HTLV-I is resistant to inactivation by human serum (Hoshin et al., Nature, Vol. 310, pgs. 324-325 (1984)). Thus, one embodiment, there is also provided a retroviral vect particle wherein a portion of the pl5E protein has be deleted and replaced with a portion of another vir protein, such as a portion of the p21 protein. p21 protein (as shown in the accompanying sequenc listing) is a protein having 176 amino acid residues, an which, in relation to pl5E, has significant amino aci sequence homology. In one embodiment, at least amino aci residues 39 to 61, and 101 to 123 are deleted from pl5 protein, and replaced with amino acid residues 34 to 56 an 96 to 118 of p21 protein. In one alternative, at leas amino acid residues 39 to 123 of pl5E protein are delete and replaced with amino acid residues 34 to 118 of p2 protein.
In another embodiment, amino acid residues 39 to 69 o pl5E protein are deleted and replaced with amino aci residues 34 to 64 of p21 protein, and amino acid residues 9 to 123 of pl5E protein are deleted and replaced with amin acid residues 91 to 118 of p21 protein.
Vector particles thus generated, and which ar resistant to inactivation by human serum, may be engineere such that the vector particles may, when introduced into patient, travel directly to a target cell or tissue. Thus in a preferred embodiment, the vector particle furthe includes a protein which contains a receptor binding regio that binds to a receptor of a human target cell, such as for example, but not limited to, the amphotropic cel receptor.
The retroviral vectors hereinabove described, may b constructed by genetic engineering techniques known to thos skilled in the art.
In one embodiment, the retroviral vector may be of th LN series of vectors, as described in Bender, et al. J.Virol., Vol. 61, pgs. 1639-1649 (1987) and Miller, et al. Biotechniques, Vol. 7, pgs. 980-990 (1989).
In another embodiment, the retroviral vector includes multiple restriction enzyme site, or multiple cloning site The multiple cloning site includes at least four cloning, o restriction enzyme sites, wherein at least two of the site have an average frequency of appearance in eukaryotic gene of less than once in 10,000 base pairs; i.e., th restriction product has an average size of at least 10,00 base pairs.
In general, such restriction sites, also sometime hereinafter referred to as "rare" sites, which have a average frequency of appearance in eukaryotic genes of les than once in 10,000 base pairs, contain a CG doublet withi their recognition sequence, such doublet appearin particularly infrequently in the mammalian genome. Anothe measure of rarity or scarcity of a restriction enzyme sit in mammals is its representation in mammalian viruses, suc as SV40. In general, an enzyme whose recognition sequenc is absent in SV40 may be a candidate for being a "rare mammalian cutter.
Examples of restriction enzyme sites having an averag frequency of appearance in eukaryotic genes of less tha once in 10,000 base pairs include, but are not limited t the NotI, SnaBI, Sail, Xhol, Clal, Sad, EagI, and Sma sites. Preferred cloning sites are selected from the grou consisting of NotI, SnaBI, Sail, and Xhol.
Preferably, the multiple cloning site has a length n greater than about 70 base pairs, and preferably no greate than about 60 base pairs. In general, the multipl restriction enzyme site, or multiple cloning site is locate between the 5'LTR and 3' TR of the retroviral vector. Th 5' end of the multiple cloning site is no greater than abou 895 base pairs from the 3' end of the 5' LTR, and preferabl at least about 375 base pairs from the 3' end of the 5' LTR The 3* end of the multiple cloning site is no greater tha about 40 base pairs fro... the 51 end of the 3' LTR, an preferably at least 11 base pairs from the 5' end of the 3 LTR.
Such vectors may be engineered from existing retrovira vectors through genetic engineering techniques known in th art such that the resulting retroviral vector includes a least four cloning sites wherein at least two of the clonin sites are selected from the group consisting of the NotI SnaBI, Sail, and Xhol cloning sites. In a preferre embodiment, the retroviral vector includes each of the NotI SnaBI, Sail, and Xhol cloning sites.
Such a retroviral vector may serve as part of a clonin system for the transfer of genes to eukaryotic cells. Thus there may be provided a cloning system for the manipulatio of genes in a retroviral vector which includes a retrovira vector including a multiple cloning site of the typ hereinabove described, and a shuttle cloning vector whic includes at least two cloning sites which are compatibl with at least two cloning sites selected from the grou consisting of NotI, SnaBI, Sail, and Xhol located on th retroviral vector. The shuttle cloning vector also include at least one desired gene which is capable of bein transferred from said shuttle cloning vector to sai retroviral vector.
The shuttle cloning vector may be constructed from basic "backbone" vector or fragment to which are ligated on or more linkers which include cloning or restriction enzym recognition sites. Included in the cloning sites are th compatible, or complementary cloning sites hereinabov described. Genes and/or promoters having ends correspondin to the restriction sites of the shuttle vector may b ligated into the shuttle vector through techniques known i the art. The shuttle cloning vector can be employed to amplif DNA sequences in prokaryotic systems. The shuttle cloni vector may be prepared from plasmids generally used i prokaryotic systems and in particular in bacteria. Thus for example, the shuttle cloning vector may be derived fro plasmids such as pBR322; pUClθ; etc.
It is also contemplated that within the scope of t present invention that the DNA encoding pl5E protein whic has been mutated to render a vector particle resistant t inactivation by human serum, may be contained in a expression vehicle other than a retroviral vector. Suc expression vehicles include, for example, viral vecto other than retroviral vectors, or any expression plasmi which is capable of being transferred into a cell line whic is capable of producing vector particles which include t mutated pl5E protein.
Such vectors or expression vehicles which contain D encoding a mutated env protein such as the mutated pl5 protein hereinabove described, are transferred into pre-packaging cell line to generate vector particles. I general, the pre-packaging cell line contains the gag an pol proteins of the virus, plus a retroviral vector lackin the structural gag, pol, and env proteins. An example o such a pre-packaging cell line is the GPL pre-packaging cel line which consists of an NIH 3T3 mouse fibroblast cell li which contains an expression plasmid for MoMuLV gag-po protein as well as the retroviral vector LNL6 (Miller, al., Biotechniques, Vol. 7, pgs. 980-990 (1989)). It is be understood, however, that the scope of the prese invention is not to be limited to any particul pre-packaging cell line.
Upon transfection of the pre-packaging cell line wi an expression vehicle containing DNA encoding a mutated e protein, the pre-packaging cell line will generate vector particles. The vector particles are then tested for complement resistance. The vector particles which are shown to be complement resistant (i.e., not inactivated by human serum), therefore, contain complement resistant envelope proteins encoded by a specific envelope expression vehicle. Such an expression vehicle can then be used, by techniques known to those skilled in the art, to produce a packaging cell line which contains an expression vehicle encoding the retroviral gag and pol proteins, and an expression vehicle containing a gene encoding the mutated env protein (such as, for example, an expression vehicle or expression plasmid containing a mutated pl5E protein such as hereinabove described), whereby such packaging cell line may be employed to generate vector particles which are resistant to inactivation by human serum. In particular, a retroviral vector which lacks the structural gag, pol, and env genes, but includes a desired gene of interest, may be transferred into such a packaging cell line. Thus, the packaging cell line may generate vector particles which contain a desired gene(s) of interest, and which are resistant to inactivation by human serum.
The vector particles generated from the packaging cell line will not be inactivated when contacted with human serum; and in addition, such vector particles, when engineered with protein containing a receptor binding region for a human receptor, are targetable, whereby the receptor binding region for a human receptor enables the vector particles to bind to a target cell. Thus, such retroviral vector particles may be directly introduced into the body (e.g., by intravenous, intramuscular, or subcutaneous injection, intranasally, orally, rectally or vaginally), and travel to a desired target cell. Such vector particles, therefore, are useful for the introduction of desire heterologous genes into target cells in vivo as a gen therapy procedure.
Thus, preferably, the vectors of the present inventio further include at least one heterologous gene Heterologous or foreign genes which may be placed into th vector or vector particles include, but are not limited to genes which encode cytokines or cellular growth factors such as lymphokines, which are growth factors fo lymphocytes. Other examples of foreign genes include, bu are not limited to, genes encoding Factor VIII, Factor IX tumor necrosis factors (TNF's), ADA, ApoE, ApoC, and Protei C.
The vectors of the present invention include one o more promoters. Suitable promoters which may be employe include, but are not limited to, the retroviral LTR; th SV40 promoter; and the human cytomegalovirus (CMV) promote described in Miller, et al., Biotechniques, Vol. 7, No. 9 pgs. 980-990 (1989), or any other promoter (eg., cellula promoters such as eukaryotic cellular promoters including but not limited to, the histone, pol III, and β-acti promoters). Other viral promoters which may be employe include, but are not limited to adenovirus promoters, T promoters, and B19 parvovirus promoters. The selection of suitable promoter will be apparent to those skilled in th art from the teachings contained herein.
The vectors of the present invention may contai regulatory elements, where necessary, to ensure tissu specific expression of the desired heterologous gene(s) and/or to regulate expression of the heterologous gene(s) i response to cellular or metabolic signals.
Although the invention has been described with respec to retroviral vector particles, other viral vector particle (such as, for example, adenovirus and adeno-associated vir particles), or synthetic particles may be construct wherein a region of the envelope protein in the vect particle may be mutated such that the vector partic becomes resistant to inactivation by human serum, there making such vector particles suitable for in vi administration.
The invention will now be described with respect to t following example; however, the scope of the prese invention is not intended to be limited thereby.
Example
Plasmid pCE2 was constructed from pBR322 such that t resulting plasmid pCE2 includes genes encoding the envelo proteins gp70 and pl5E. pBR322 (Figure 1) was cut wi EcoRI and filled in to destroy the EcoRI site to gi pBR322Z Rl. pBR3224Rl was then cut with Ndel and filled to destroy the Ndel site to give pBR322 A R pBR322_4 R N was digested with Hindi11 and EcoRV, a cloned into the Hindlll/EcoRV fragment was a Hindlll/Fs cassette containing the gp70 and pl5E genes under t control of a cytomegalovirus (CMV) intermediate ear promoter with a polyA (adenine) tail from SV40 (Figure from plasmid pCEE. (Figure 3). The Hindlll/Fspl casset obtained from plasmid pCEE contains a CMV intermediate ear promoter in which the Ball/Sac11 (bp 21 to bp 766) w converted to an Hindlll/Sall fragment by linker additio the ecotropic envelope Bglll/Nhel fragment (bp 5408 to 7847 of MoMuLV, encoding gp70 and pl5E) was filled and Eco linkers were added; and the SV40 poly A signal from Bell BamHI (bp 2770 to bp 2533) was cloned into a BamHI si (thereby destroying the Bell site). A Bglll site was add at the 31 end of the gp70 gene. (This addition does n alter any amino acids). The resulting plasmid is pCE (Figure 4. )
To create the mutations in the pl5E gene, sub-loning i carried out in a different plasmid called pUC-E2 (Figure 6.) This plasmid is pUC18 (Figure 5) with the PvuI fragment removed and replaced with EcoRI linkers. Such wa accomplished by digesting pUC18 with PvuII to remove a 32 bp PvuII fragment, and EcoRI linkers were then added. Int the EcoRI site was cloned the MoMuLV ecotropic envelope gen (i.e., the gp70 and pl5E genes from pCE2) from the Bgll site (5408) to the Nhel site (7847), which have been blunte and had EcoRI linkers added. The resulting pUC-E2 plasmi (Figure 6) therefore has unique Bglll, Spel, Clal, and PvuI sites in and around the pl5E gene.
PCR primers are then synthesized to encode th following mrtations in the pl5E protein (using pCE2 as template):
1. Amino acid residue 117 is changed from Arg to Glu and amino acid residue 122 is changed from Glu to Gin;
2. Amino acid residue 104 is changed from Arg to His amino acid residue 105 is changed from Asp to Asn, amin acid residue 109 is changed from Lys to Gin, and amino aci residue 111 is changed from Arg to Gin; and
3. Amino acid residue 104 is changed from Arg to His amino acid residue 105 is changed from Asp to Asn, amin acid residue 109 is changed from Lys to Gin, amino aci residue 111 is hanged from Arg to Gin, amino acid residu 117 is changed from Arg to Glu, and amino acid residue 12 is changed from Glu to Gin.
Each PCR product is digested with Spel and PvuII, a cloned into pUC-E2 at the unique Spel and PvuII sites. T resulting plasmids are then sequenced to confirm the poi mutations. DNA fragments bearing these mutations are the subcloned into the expression plasmid pCE2. pCE2 i digested with EcoRI and the envelope DNA fragment is remove and replaced with the EcoRI envelope fragment from th pUC-E2 plasmids. The resulting pCE2 plasmids are the checked for orientation of the EcoRI fragment and sequence again (only at the cloning site junctions and at the region bearing the point mutations) to confirm the presence of th newly created mutated pl5E genes. The resulting expressio plasmids are identified as follows: pCR68 - includes mutations in which amino acid residu 117 is changed from Arg to Glu, and amino acid residu 122 is changed from Glu to Gin; pCR69 - includes mutations in which amino acid residu 104 is changed from Arg to His, amino acid residue 10 is changed from Asp to Asn, amino acid residue 109 i changed from Lys to Gin, and amino acid residue 111 i changed from Arg to Gin; and pCR70 - includes mutations in which amino acid residu 104 is changed from Arg to His, amino acid residue 10 is changed from Asp to Asn, amino acid residue 109 i changed from Lys to Gin, amino acid residue 111 i changed from Arg to Gin, amino acid residue 117 i changed from Arg to Glu, and amino acid residue 122 i changed from Glu to Gin.
Plasmids pCR68, pCR69, and pCR70 are transfecte separately into the GPL pre-packaging cell line. The GP pre-packaging cell line consists of an NIH 3T3 mous fibroblast cell line which contains an expression plasmi for MoMuLV gag-pol protein as well as the retroviral vecto LNL6 (Miller, et al., 1989). Upon transfection with pCR68 pCR69, or pCR70, the GPL packaging cell line produces vecto particles. Transiently expressed vector particles are collecte with cell supernatant at 48-72 hrs. post-transfection.
The vector particles generated as hereinabove describ may then be assayed for vector titer by techniques known those skilled in the art. The vector particles may also collected in viral supernatant and concentrated, i necessary, according to procedures known to those skilled i the art in order to employ such vector particles in assa or in therapeutic procedures.
Advantages of the present invention include the abili to introduce vector particles directly into a human patie whereby the vector particle is not lysed or inactivated human serum upon such introduction. Thus, the vect particles of the present invention enable one to deliv desired genes to a patient in vivo. Such vector particl may also be engineered such that they are "targetable", well as injectable, thereby enabling the vector particles travel directly to a target cell or tissue without bei lysed or inactivated by human serum.
It is to be understood, however, that the scope of t present invention is not to be limited to the specifi embodiments described above. The invention may be practic other than as particularly described and still be within t scope of the accompanying claims.
SEQUENCE LISTING (1) GENERAL INFORMATION:
(i) APPLICANT: Anderson, W. French
Mason, James M. (ii) TITLE OF INVENTION: Vector Particles
Resistant to Inactivatio by Human Serum (iii) NUMBER OF SEQUENCES: 2 (iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Carella, Byrne, Bain,
Gilfillan, Cecchi & Stewart
(B) STREET: 6 Becker Farm Road
(C) CITY: Roseland
(D) STATE: New Jersey
(E) COUNTRY: USA
(F) ZIP: 07068 (v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: 3.5 inch diskette
(B) COMPUTER: IBM PS/2
(C) OPERATING SYSTEM: PC-DOS
(D) SOFTWARE: DW4.V2 (vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION: (Vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(Viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Olεtein, Elliot M.
(B) REGISTRATION NUMBER: 24,025
(C) REFERENCE/DOCKET NUMBER: 271010-73 (ix) TELECOMMUNICATION INFORMATION: (A) TELEPHONE: 201-994-1700
(B) TELEFAX: 201-994-1744
(2) INFORMATION FOR SEQ ID NO: 1:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 196 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (ix) FEATURE:
(A) NAME/KEY: pl5E protein, (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1:
Glu Pro Val Ser Leu Thr Leu Ala Leu Leu
5 10
Leu Gly Gly Leu Thr Met Gly Gly lie Ala
15 20
Ala Gly lie Gly Thr Gly Thr Thr Ala Leu
25 30
Met Ala Thr Gin Gin Phe Gin Gin Leu Gin
35 40
Ala Ala Val Gin Asp Asp Leu Arg Glu Val
45 50
Glu Lys Ser lie Ser Asn Leu Glu Lys Ser
55 60
Leu Thr Ser Leu Ser Glu Val Val Leu Gin
65 70
Asn Arg Arg Gly Leu Asp Leu Leu Phe Leu
75 80
Lys Glu Gly Gly Leu Cyε Ala Ala Leu Lys
85 90
Glu Glu Cys Cys Phe Tyr Ala Asp His Thr
95 100 Gly Leu Val Arg Asp Ser Met Ala Lys Leu
105 110
Arg Glu Arg Leu Asn Gin Arg Gin Lys Leu
115 120
Phe Glu Ser Thr Gin Gly Trp Phe Glu Gly
125 130
Leu Phe Asn Arg Ser Pro Trp Phe Thr Thr
135 140
Leu lie Ser Thr lie Met Gly Pro Leu lie
145 150
Val Leu Leu Met lie Leu Leu Phe Gly Pro
155 160
Cys lie Leu Asn Arg Leu Val Gin Phe Val
165 170
Lys Asp Arg lie Ser Val Val Gin Ala Leu
175 180
Val Leu Thr Gin Gin Tyr His Gin Leu Lys
185 190
Pro lie Glu Tyr Glu Pro
195
(2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 176 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS:
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(ix) FEATURE
(A) NAME/KEY: p21 protein (Xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Ala Val Pro Val Ala Val Trp Leu Val Ser
5 10
Ala Leu Ala Met Gly Ala Gly Val Ala Gly
15 20
Arg lie Thr Gly Ser Met Ser Leu Ala Ser
25 30
Gly Lys Ser Leu Leu His Glu Val Asp Lys
35 40
Asp lie Ser Gin Leu Thr Gin Ala lie Val
45 50
Lys Asn His Lys Asn Leu Leu Lys lie Ala
55 60
Gin Tyr Ala Ala Gin Asn Arg Arg Gly Leu
65 70
Asp Leu Leu Phe Trp Glu Gin Gly Gly Leu
75 80
Cyε Lys Ala Leu Gin Glu Gin Cys Cys Phe
85 90
Leu Asn lie Thr Asn Ser His Val Ser lie
95 100
Leu Gin Glu Arg Pro Pro Leu Glu Asn Arg
105 110
Val Leu Thr Gly Trp Gly Leu Asn Trp Asp
115 120
Leu Gly Leu Ser Gin Trp Ala Arg Glu Ala
125 130
Leu Gin Thr Gly lie Thr Leu Val Ala Leu
135 140
Leu Leu Leu Val lie Leu Ala Gly Pro Cys
145 150 lie Leu Arg Gin Leu Arg His Leu Pro Ser
155 160 Arg Val Arg Tyr Pro Hiε Tyr -Ser Leu lie
165 170
Asn Pro Glu Ser Ser Leu
175

Claims

WHAT IS CLAIMED IS:
1. A retroviral vector particle, said vect particle being resistant to inactivation by human serum.
2. The vector particle of Claim 1 wherein sa vector particle includes pl5E protein, and wherein a porti but not all of the pl5E protein has been mutated to rend the vector particles reεistant to inactivation by hum serum.
3. The vector particle of Claim 2 wherein portion of the pl5E protein has been mutated such that least one amino acid of pl5E protein has been changed.
4. The vector particle of Claim 3 wherein at lea one portion of a complement binding region of pl5E prote has been mutated.
5. The vector particle of Claim 4 wherein sa complement binding region is selected from the gro consisting of amino acid residues 39 to 61 and 101 to 123 pl5E protein.
6. The vector particle of Claim 5 wherein sa complement binding region is amino acid residues 101 to 1 of pl5E protein.
7. The vector particle of Claim 6 wherein pl protein is mutated such that amino acid residue 122 of pl protein is changed.
8. The vector particle of Claim 6 wherein pl protein is mutated such that amino acid residues 117 and 1 are changed.
9. The vector particle of Claim 8 wherein pl protein is mutated such that amino acid residue 117 changed from Arg to Glu, and amino acid residue 122 changed from Glu to Gin.
10. The vector particle of Claim 6 wherein pl5 protein is mutated such that amino acid residues 104, 105 109, and 111 are changed.
11. The vector particle of Claim 10 wherein pl5 protein is mutated such that amino acid residue 104 i changed from Arg to His, amino acid residue 105 is chang from Asp to Asn, amino acid residue 109 is changed from L to Gin, and amino acid residue 111 is changed from Arg t Gin.
12. The vector particle of Claim 6 wherein pl5 protein is mutated such that amino acid residues 104, 105 109, 111, 117, and 122 are changed.
13. The vector particle of Claim 12 wherein pl protein is mutated such that amino acid residue 104 i changed from Arg to His, amino acid residue 105 is chang from Asp to Asn, amino acid residue 109 is changed from L to Gin, amino acid residue 111 is changed from Arg to Gi amino acid residue 117 is changed from Arg to Glu, and ami acid residue 122 is changed from Glu to Gin.
14. The vector particle of Claim 5 wherein pl5 protein is mutated such that at least one positively o negatively charged amino acid is changed to an amino aci having the opposite charge.
15. The vector particle of Claim 5 wherein pl5 protein is mutated such that at least one positively charg or negatively charged amino acid is changed to a non-charg amino acid.
16. The vector particle of Claim 1 wherein sai vector particle further includes a protein containing receptor binding region that binds to a receptor of a hum target cell.
17. The vector particle of Claim 16 and furthe containing a heterologous gene.
18. Eukaryotic cells transformed with the vecto particles of Claim 17.
19. An expression vehicle including DNA encodin pl5E protein, wherein said DNA encoding pl5E protein i mutated such that when said mutated pl5E protein encoded b said DNA is included in a vector particle, said vecto particle is resistant to inactivation by human serum.
20. A packaging cell line including the expreεsio vehicle of Claim 19.
21. A viral vector particle, said viral vecto particle being resistant to inactivation by human serum.
EP93913964A 1992-06-10 1993-05-14 Vector particles resistant to inactivation by human serum. Withdrawn EP0644946A4 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US896603 1986-08-13
US89660392A 1992-06-10 1992-06-10
PCT/US1993/004706 WO1993025698A1 (en) 1992-06-10 1993-05-14 Vector particles resistant to inactivation by human serum

Publications (2)

Publication Number Publication Date
EP0644946A1 true EP0644946A1 (en) 1995-03-29
EP0644946A4 EP0644946A4 (en) 1997-03-12

Family

ID=25406489

Family Applications (1)

Application Number Title Priority Date Filing Date
EP93913964A Withdrawn EP0644946A4 (en) 1992-06-10 1993-05-14 Vector particles resistant to inactivation by human serum.

Country Status (4)

Country Link
EP (1) EP0644946A4 (en)
JP (1) JPH09507741A (en)
CA (1) CA2137361A1 (en)
WO (1) WO1993025698A1 (en)

Families Citing this family (227)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU7516594A (en) * 1993-07-28 1995-02-28 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Pre-binding of retroviral vector particles with complement components to enable the performance of human gene therapy (in vivo)
ATE287732T1 (en) * 1994-08-17 2005-02-15 Genetic Therapy Inc RETROVIRAL VECTORS PRODUCED BY PRODUCTION CELL LINES RESISTANT TO LYSIS BY HUMAN SERUM
US6818439B1 (en) 1994-12-30 2004-11-16 Chiron Corporation Methods for administration of recombinant gene delivery vehicles for treatment of hemophilia and other disorders
GB9517263D0 (en) * 1995-08-23 1995-10-25 Cancer Res Campaign Tech Expression systems
GB9525639D0 (en) 1995-12-15 1996-02-14 Isis Innovation Improved retroviral vectors
EP0951544A2 (en) * 1996-07-03 1999-10-27 Chiron Corporation Methods for administration of recombinant gene delivery vehicles for treatment of human disease
EP1860192A1 (en) 1996-09-17 2007-11-28 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating intracellular diseases
US6544523B1 (en) 1996-11-13 2003-04-08 Chiron Corporation Mutant forms of Fas ligand and uses thereof
DE69838584T2 (en) 1997-08-04 2008-06-26 Cell Genesys, Inc., Foster City ENHANCERS OF HUMAN GLANDULAR CALLIQUE, VECTORS CONTAINING HIM, AND METHODS FOR ITS USE
WO1999019466A2 (en) 1997-10-14 1999-04-22 Darwin Molecular Corporation Thymidine kinase mutants and fusion proteins having thymidine kinase and guanylate kinase activities
DE69841807D1 (en) 1997-11-06 2010-09-16 Novartis Vaccines & Diagnostic NEISSERIAL ANTIGENE
DE69941567D1 (en) 1998-01-14 2009-12-03 Novartis Vaccines & Diagnostic ANTIGENE FROM NEISSERIA MENINGITIDIS
CN1210305C (en) 1998-05-01 2005-07-13 希龙公司 Neisseria meningitidis antigens and compositions
EP1121437B1 (en) 1998-10-15 2008-02-20 Novartis Vaccines and Diagnostics, Inc. Metastatic breast and colon cancer regulated genes
IL143266A0 (en) 1998-11-27 2002-04-21 Darwin Discovery Ltd Transforming growth factor-beta binding proteins and pharmaceutical compositions for increasing bone mineral content utilizing the same
US20040009535A1 (en) 1998-11-27 2004-01-15 Celltech R&D, Inc. Compositions and methods for increasing bone mineralization
AU2367600A (en) 1998-12-16 2000-07-03 Chiron Corporation Human cyclin-dependent kinase ((hpnqalre))
US7063850B1 (en) 1998-12-22 2006-06-20 University Of Tennessee Research Foundation Protective antigen of group A Streptococci
CA2360347C (en) 1998-12-31 2013-05-07 Chiron Corporation Improved expression of hiv polypeptides and production of virus-like particles
US7935805B1 (en) 1998-12-31 2011-05-03 Novartis Vaccines & Diagnostics, Inc Polynucleotides encoding antigenic HIV Type C polypeptides, polypeptides and uses thereof
US6911429B2 (en) 1999-04-01 2005-06-28 Transition Therapeutics Inc. Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US6864235B1 (en) 1999-04-01 2005-03-08 Eva A. Turley Compositions and methods for treating cellular response to injury and other proliferating cell disorders regulated by hyaladherin and hyaluronans
US7368261B1 (en) 1999-04-30 2008-05-06 Novartis Vaccines And Diagnostics Srl Conserved Neisserial antigens
GB9911683D0 (en) 1999-05-19 1999-07-21 Chiron Spa Antigenic peptides
GB9916529D0 (en) 1999-07-14 1999-09-15 Chiron Spa Antigenic peptides
EP2275552B1 (en) 1999-10-29 2015-09-09 GlaxoSmithKline Biologicals SA Neisserial antigenic peptides
JP2003516731A (en) 1999-11-18 2003-05-20 カイロン コーポレイション Human FGF-21 gene and gene expression product
DK1248647T3 (en) 2000-01-17 2010-09-27 Novartis Vaccines & Diagnostic Outer membrane vesicle (OMV) vaccine comprising N. meningitidis serogroup B outer membrane proteins
EP1854476A3 (en) 2000-02-09 2008-05-07 Bas Medical, Inc. Use of relaxin to treat diseases related to vasoconstriction
US20020082205A1 (en) 2000-03-08 2002-06-27 Nobuyuki Itoh Human FGF-23 gene and gene expression products
EP1950297A2 (en) 2000-05-31 2008-07-30 Novartis Vaccines and Diagnostics, Inc. Compositions and methods for treating neoplastic disease using chemotherapy and radiation sensitizers
US7700359B2 (en) 2000-06-02 2010-04-20 Novartis Vaccines And Diagnostics, Inc. Gene products differentially expressed in cancerous cells
PT1370684E (en) 2000-06-15 2008-09-08 Novartis Vaccines & Diagnostic Polynucleotides related to colon cancer
SG165981A1 (en) 2000-10-27 2010-11-29 Chiron Srl Nucleic acids and proteins from streptococcus groups a & b
AU2002227365A1 (en) 2000-12-07 2002-06-18 Chiron Corporation Endogenous retroviruses up-regulated in prostate cancer
US7829084B2 (en) 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
CN1911965B (en) 2001-01-17 2013-05-29 新兴产品开发西雅图有限公司 Binding domain-immunoglobulin fusion proteins
GB0107661D0 (en) 2001-03-27 2001-05-16 Chiron Spa Staphylococcus aureus
GB0107658D0 (en) 2001-03-27 2001-05-16 Chiron Spa Streptococcus pneumoniae
AU2002258728A1 (en) 2001-04-06 2002-10-21 Georgetown University Gene brcc-3 and diagnostic and therapeutic uses thereof
AU2002305151A1 (en) 2001-04-06 2002-10-21 Georgetown University Gene scc-112 and diagnostic and therapeutic uses thereof
WO2002081639A2 (en) 2001-04-06 2002-10-17 Georgetown University Gene brcc2 and diagnostic and therapeutic uses thereof
DE60228477D1 (en) 2001-05-08 2008-10-02 Darwin Molecular Corp PROCESS FOR REGULATING THE IMMUNE FUNCTION IN PRIMATES USING THE FOXP3 PROTEIN
US7211659B2 (en) 2001-07-05 2007-05-01 Chiron Corporation Polynucleotides encoding antigenic HIV type C polypeptides, polypeptides and uses thereof
EP2292772A1 (en) 2001-07-05 2011-03-09 Novartis Vaccines and Diagnostics, Inc. HIV vaccination with a DNA encoding a HIV polypeptide and a HIV polypeptide
US6939959B2 (en) 2001-10-26 2005-09-06 Id Biomedical Corporation Of Washington Efficient protein expression system
US7498407B2 (en) 2001-11-09 2009-03-03 Georgetown University Vascular endothelial cell growth inhibitor, VEGI-192a
CN100515494C (en) 2001-12-12 2009-07-22 启龙有限公司 Immunisation against chlamydia trachomatis
ES2323456T3 (en) 2002-01-08 2009-07-16 Novartis Vaccines And Diagnostics, Inc. GENETIC PRODUCTS DIFFERENTIALLY EXPRESSED IN CANCEROSE CHEST CELLS AND THEIR METHODS OF USE.
IL163988A0 (en) 2002-03-15 2005-12-18 Wyeth Corp Mutants of the p4 protein of nontypable haemophilus influenzae with reduced enzymatic activity
EP2093233A1 (en) 2002-03-21 2009-08-26 Sagres Discovery, Inc. Novel compositions and methods in cancer
US7244565B2 (en) 2002-04-10 2007-07-17 Georgetown University Gene shinc-3 and diagnostic and therapeutic uses thereof
AU2003276679A1 (en) 2002-06-13 2003-12-31 Chiron Corporation Vectors for expression of hml-2 polypeptides
UA80447C2 (en) 2002-10-08 2007-09-25 Methods for treating pain by administering nerve growth factor antagonist and opioid analgesic
US7569364B2 (en) 2002-12-24 2009-08-04 Pfizer Inc. Anti-NGF antibodies and methods using same
US9498530B2 (en) 2002-12-24 2016-11-22 Rinat Neuroscience Corp. Methods for treating osteoarthritis pain by administering a nerve growth factor antagonist and compositions containing the same
NZ587852A (en) 2002-12-24 2012-02-24 Rinat Neuroscience Corp Anti-NGF antibodies and methods using same
US20040170982A1 (en) 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
EP1592708A2 (en) 2003-02-14 2005-11-09 Sagres Discovery, Inc. Therapeutic gpcr targets in cancer
US7767387B2 (en) 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
WO2004073653A2 (en) 2003-02-19 2004-09-02 Rinat Neuroscience Corp. Methods for treating pain by administering a nerve growth factor antagonist and an nsaid and compositions containing the same
GB0308198D0 (en) 2003-04-09 2003-05-14 Chiron Srl ADP-ribosylating bacterial toxin
US20070178066A1 (en) 2003-04-21 2007-08-02 Hall Frederick L Pathotropic targeted gene delivery system for cancer and other disorders
EP1619951B1 (en) 2003-04-21 2011-06-22 Epeius Biotechnologies Corporation Methods and compositions for treating disorders
WO2005020928A2 (en) 2003-08-29 2005-03-10 The Regents Of The University Of California Agents and methods for enhancing bone formation by oxysterols in combination with bone morphogenic proteins
EP2402756A3 (en) 2003-12-23 2012-02-22 Rinat Neuroscience Corp. Agonist anti-trkC antibodies and methods using same
CA2561696C (en) 2004-03-29 2013-09-03 Galpharma Co., Ltd. Novel modified galectin 9 proteins and use thereof
PT1732949E (en) 2004-04-07 2010-03-23 Univ Minnesota Methods for treating bone cancer pain by administering a nerve growth factor antagonist
ES2622394T3 (en) 2004-07-09 2017-07-06 The Henry M. Jackson Foundation For The Advancement Of Military Medicine, Inc. Soluble forms of hendra virus g glycoprotein
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
KR20070040824A (en) 2004-07-30 2007-04-17 리나트 뉴로사이언스 코퍼레이션 Antibodies directed against amyloid-beta peptide and methods using same
CA2598488A1 (en) 2005-02-18 2006-08-31 Novartis Vaccines And Diagnostics, Inc. Immunogens from uropathogenic escherichia coli
WO2006110599A2 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cacna1e in cancer diagnosis, detection and treatment
EP2083088A3 (en) 2005-04-07 2009-10-14 Novartis Vaccines and Diagnostics, Inc. Cancer-related genes
KR20080039929A (en) 2005-07-22 2008-05-07 와이스 테라퓨틱스 가부시키가이샤 Anti-cd26 antibodies and methods of use thereof
DK1912675T3 (en) 2005-07-25 2014-03-24 Emergent Product Dev Seattle B-cell reduction using specific and cd37-cd20-specific binding molecules
RS20080200A (en) 2005-11-14 2009-07-15 Rinat Neuroscience Corp., Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
JP2009525319A (en) 2006-02-02 2009-07-09 ライナット ニューロサイエンス コーポレイション Method of treating obesity by administering a trkB antagonist
US9670244B2 (en) 2006-02-27 2017-06-06 The Regents Of The University Of California Oxysterol compounds and the hedgehog pathway
SG172684A1 (en) 2006-06-07 2011-07-28 Bioalliance Cv Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
EP2035035A2 (en) 2006-06-09 2009-03-18 Novartis AG Immunogenic compositions for streptococcus agalactiae
CN105837690A (en) 2006-06-12 2016-08-10 新兴产品开发西雅图有限公司 Single-chain multivalent binding proteins with effector function
JP2010500399A (en) 2006-08-16 2010-01-07 ノバルティス アーゲー Immunogen from Urinary Pathogenic Escherichia coli
CN101516396B (en) 2006-09-26 2013-10-09 传染性疾病研究院 Vaccine composition containing synthetic adjuvant
US20090181078A1 (en) 2006-09-26 2009-07-16 Infectious Disease Research Institute Vaccine composition containing synthetic adjuvant
EP2139447A2 (en) 2007-03-20 2010-01-06 Harold Brem Gm-csf cosmeceutical compositions and methods of use thereof
AP2009005028A0 (en) 2007-04-10 2009-12-31 Univ Tulane Soluble and membrane-anchored forms of lassa virussubunit proteins
GB0714963D0 (en) 2007-08-01 2007-09-12 Novartis Ag Compositions comprising antigens
US9526737B2 (en) 2007-12-03 2016-12-27 The Regents Of The University Of California Oxysterols for activation of hedgehog signaling, osteoinduction, antiadipogenesis, and Wnt signaling
JP5737944B2 (en) 2007-12-17 2015-06-17 ファイザー・リミテッドPfizer Limited Treatment of interstitial cystitis
CN105732813A (en) 2007-12-18 2016-07-06 生物联合公司 Antibodies recognizing a carbohydrate containing epitope on cd-43 and cea expressed on cancer cells and methods using same
EP3023502A1 (en) 2008-04-10 2016-05-25 Cell Signaling Technology, Inc. Compositions and methods for detecting egfr mutations in cancer
JP6013733B2 (en) 2008-04-11 2016-10-25 エマージェント プロダクト デベロップメント シアトル, エルエルシー CD37 immunotherapeutic and its combination with bifunctional chemotherapeutics
WO2009150623A1 (en) 2008-06-13 2009-12-17 Pfizer Inc Treatment of chronic prostatitis
TWI516501B (en) 2008-09-12 2016-01-11 禮納特神經系統科學公司 Pcsk9 antagonists
WO2010039536A2 (en) 2008-09-23 2010-04-08 President And Fellows Of Harvard College Sirt4 and uses thereof
US9181315B2 (en) 2009-01-08 2015-11-10 Dana-Farber Cancer Institute, Inc. Compositions and methods for induced brown fat differentiation
WO2010086828A2 (en) 2009-02-02 2010-08-05 Rinat Neuroscience Corporation Agonist anti-trkb monoclonal antibodies
EP2403526B1 (en) 2009-03-06 2019-05-15 GlaxoSmithKline Biologicals SA Chlamydia antigens
WO2010118243A2 (en) 2009-04-08 2010-10-14 Genentech, Inc. Use of il-27 antagonists to treat lupus
EP2419129A2 (en) 2009-04-14 2012-02-22 Novartis AG Compositions for immunising against staphylococcus aerus
WO2010141861A1 (en) 2009-06-05 2010-12-09 Infectious Disease Research Institute Synthetic glucopyranosyl lipid adjuvants
WO2010146511A1 (en) 2009-06-17 2010-12-23 Pfizer Limited Treatment of overactive bladder
US20110135674A1 (en) * 2009-06-30 2011-06-09 Abbott Laboratories Markers of XMRV Infection and Uses Thereof
ES2526996T3 (en) 2009-07-16 2015-01-19 Novartis Ag Detoxified immunogens from Escherichia coli
WO2011038063A1 (en) 2009-09-28 2011-03-31 The Trustees Of The University Of Pennsylvania Method of diagnosing and treating interstitial cystitis
GB0919690D0 (en) 2009-11-10 2009-12-23 Guy S And St Thomas S Nhs Foun compositions for immunising against staphylococcus aureus
JP2013517772A (en) 2010-01-21 2013-05-20 デイナ ファーバー キャンサー インスティチュート,インコーポレイテッド A situation-specific gene screening platform to aid gene discovery and target confirmation
US8298535B2 (en) 2010-02-24 2012-10-30 Rinat Neuroscience Corp. Anti-IL-7 receptor antibodies
GB201003333D0 (en) 2010-02-26 2010-04-14 Novartis Ag Immunogenic proteins and compositions
RU2570729C2 (en) 2010-03-11 2015-12-10 Ринат Ньюросайенс Корпорейшн Antibodies with ph-dependent antigen binding
GB201005625D0 (en) 2010-04-01 2010-05-19 Novartis Ag Immunogenic proteins and compositions
WO2011133931A1 (en) 2010-04-22 2011-10-27 Genentech, Inc. Use of il-27 antagonists for treating inflammatory bowel disease
US8747844B2 (en) 2010-07-30 2014-06-10 Saint Louis University Methods of treating pain
WO2012064743A2 (en) 2010-11-08 2012-05-18 The Johns Hopkins University Methods for improving heart function
WO2012072769A1 (en) 2010-12-01 2012-06-07 Novartis Ag Pneumococcal rrgb epitopes and clade combinations
US9198975B2 (en) 2010-12-01 2015-12-01 The University Of North Carolina At Chapel Hill Methods and compositions for targeting sites of neovascular growth
JP6054942B2 (en) 2011-04-08 2016-12-27 イミューン デザイン コーポレイション Immunogenic compositions and methods of using the compositions to elicit humoral and cellular immune responses
US20130071375A1 (en) 2011-08-22 2013-03-21 Saint Louis University Compositions and methods for treating inflammation
WO2013028527A1 (en) 2011-08-23 2013-02-28 Indiana University Research And Technology Corporation Compositions and methods for treating cancer
US10093705B2 (en) 2011-09-13 2018-10-09 Dana-Farber Cancer Institute, Inc. Compositions and methods for brown fat induction and activity using FNDC5
WO2013055911A1 (en) 2011-10-14 2013-04-18 Dana-Farber Cancer Institute, Inc. Znf365/zfp365 biomarker predictive of anti-cancer response
US8871908B2 (en) 2011-11-11 2014-10-28 Rinat Neuroscience Corp. Antibodies specific for Trop-2 and their uses
WO2013093693A1 (en) 2011-12-22 2013-06-27 Rinat Neuroscience Corp. Staphylococcus aureus specific antibodies and uses thereof
KR20140102710A (en) 2011-12-22 2014-08-22 리나트 뉴로사이언스 코프. Human growth hormone receptor antagonist antibodies and methods of use thereof
EP2811981B1 (en) 2012-02-07 2019-05-08 Infectious Disease Research Institute Improved adjuvant formulations comprising tlr4 agonists and methods of using the same
PL2844282T3 (en) 2012-05-04 2019-11-29 Pfizer Prostate-associated antigens and vaccine-based immunotherapy regimens
RU2632191C2 (en) 2012-05-07 2017-10-03 Те Риджентс Оф Те Юниверсити Оф Калифорния Oxy133 OXYSTEROL ANALOGUE INDUCTS OSTEOGENESIS AND SIGNAL WAY HEDGEHOG AND INHIBITED LIPOGENESIS
BR112014028476A2 (en) 2012-05-16 2017-08-01 Immune Design Corp immunogenic fragment of a polypeptide, isolated polynucleotide, immunogenic pharmaceutical composition, methods for treating an infection, for generating an immune response in a subject, and for immunizing a subject
US8603470B1 (en) 2012-08-07 2013-12-10 National Cheng Kung University Use of IL-20 antagonists for treating liver diseases
JP2015536339A (en) 2012-11-09 2015-12-21 ファイザー・インク Platelet-derived growth factor B specific antibodies and compositions and uses thereof
WO2014153258A2 (en) 2013-03-14 2014-09-25 Epeius Biotechnologies Corporation Improved thymidine kinase gene
AU2014240045A1 (en) 2013-03-15 2015-09-10 Dyax Corp. Anti-plasma kallikrein antibodies
SG11201508092YA (en) 2013-04-18 2015-10-29 Immune Design Corp Gla monotherapy for use in cancer treatment
EP2991652A4 (en) 2013-05-02 2016-12-07 Univ California Bone-selective osteogenic oxysterol-bone targeting agents
AU2014264295A1 (en) 2013-05-07 2015-10-29 Rinat Neuroscience Corp. Anti-glucagon receptor antibodies and methods of use thereof
US9463198B2 (en) 2013-06-04 2016-10-11 Infectious Disease Research Institute Compositions and methods for reducing or preventing metastasis
WO2015006641A2 (en) 2013-07-12 2015-01-15 Georgia State University Research Foundation, Inc. Methods and compositions for interference with dna polymerase and dna synthesis
US10208125B2 (en) 2013-07-15 2019-02-19 University of Pittsburgh—of the Commonwealth System of Higher Education Anti-mucin 1 binding agents and uses thereof
MY190252A (en) 2013-08-02 2022-04-08 Pfizer Anti-cxcr4 antibodies and antibody-drug conjugates
RU2708140C1 (en) 2013-11-13 2019-12-04 Пфайзер Инк. Antibodies specific for ligand 1a, similar to tumor necrosis factor, and their compositions and application
WO2015087187A1 (en) 2013-12-10 2015-06-18 Rinat Neuroscience Corp. Anti-sclerostin antibodies
WO2015109212A1 (en) 2014-01-17 2015-07-23 Pfizer Inc. Anti-il-2 antibodies and compositions and uses thereof
HRP20240159T1 (en) 2014-03-21 2024-04-12 Teva Pharmaceuticals International Gmbh Antagonist antibodies directed against calcitonin gene-related peptide and methods using same
WO2015164743A2 (en) 2014-04-24 2015-10-29 Dana-Farber Cancer Institute, Inc. Tumor suppressor and oncogene biomarkers predictive of anti-immune checkpoint inhibitor response
WO2015168474A1 (en) 2014-04-30 2015-11-05 President And Fellows Of Harvard College Fusion proteins for treating cancer and related methods
US10329556B2 (en) 2014-05-13 2019-06-25 Bioatla, Llc Conditionally active biological proteins
US9840553B2 (en) 2014-06-28 2017-12-12 Kodiak Sciences Inc. Dual PDGF/VEGF antagonists
US11111288B2 (en) 2014-08-28 2021-09-07 Bioatla, Inc. Conditionally active chimeric antigen receptors for modified t-cells
RU2764074C2 (en) 2014-08-28 2022-01-13 Байоатла, Ллк Conditionally active chimeric antigen receptors for modified t-cells
BR112017004131A2 (en) 2014-09-03 2017-12-12 Bioatla Llc method of producing a conditionally active biological protein, conditionally active biological protein, chimeric antigen receptor, and cytotoxic cell.
SG11201701775VA (en) 2014-09-09 2017-04-27 Unum Therapeutics Chimeric receptors and uses thereof in immune therapy
US20170248603A1 (en) 2014-10-06 2017-08-31 Dana-Farber Cancer Institute, Inc. Angiopoiten-2 biomarkers predictive of anti-immune checkpoint response
CN107106876B (en) 2014-10-09 2021-06-11 丹娜法伯癌症研究院 Multiple-variable IL-2 dosage regimen for treating immune disorders
TWI595006B (en) 2014-12-09 2017-08-11 禮納特神經系統科學公司 Anti-pd-1 antibodies and methods of use thereof
WO2016144673A1 (en) 2015-03-06 2016-09-15 Dana-Farber Cancer Institute, Inc. Pd-l2 biomarkers predictive of pd-1 pathway inhibitor responses in esophagogastric cancers
US9758575B2 (en) 2015-04-06 2017-09-12 Yung Shin Pharmaceutical Industrial Co. Ltd. Antibodies which specifically bind to canine vascular endothelial growth factor and uses thereof
SI3283106T1 (en) 2015-04-13 2022-04-29 Pfizer Inc. Therapeutic antibodies and their uses
US10877045B2 (en) 2015-07-21 2020-12-29 Saint Louis University Compositions and methods for diagnosing and treating endometriosis-related infertility
IL257047B1 (en) 2015-07-21 2024-07-01 Dyax Corp A monoclonal antibody inhibitor of factor xiia
US11066481B2 (en) 2015-07-23 2021-07-20 The Regents Of The University Of California Antibodies to coagulation factor XIa and uses thereof
IL302353A (en) 2015-08-19 2023-06-01 Pfizer Tissue factor pathway inhibitor antibodies and uses thereof
SI3350220T1 (en) 2015-09-15 2021-12-31 Scholar Rock, Inc. Anti-pro/latent-myostatin antibodies and uses thereof
JP7002446B2 (en) 2015-09-21 2022-03-04 アプティーボ リサーチ アンド デベロップメント エルエルシー CD3 binding polypeptide
JP7017506B2 (en) 2015-10-16 2022-02-08 ザ・トラスティーズ・オブ・コロンビア・ユニバーシティ・イン・ザ・シティ・オブ・ニューヨーク Compositions and Methods for Inhibition of Strain-Specific Antigens
US11207393B2 (en) 2015-10-16 2021-12-28 President And Fellows Of Harvard College Regulatory T cell PD-1 modulation for regulating T cell effector immune responses
CN115636880A (en) 2015-10-23 2023-01-24 辉瑞有限公司 anti-IL-2 antibodies and compositions and uses thereof
WO2017075037A1 (en) 2015-10-27 2017-05-04 Scholar Rock, Inc. Primed growth factors and uses thereof
CA3002676A1 (en) 2015-10-29 2017-05-04 Dana-Farber Cancer Institute, Inc. Methods for identification, assessment, prevention, and treatment of metabolic disorders using pm20d1 and n-lipidated amino acids
CN106699889A (en) 2015-11-18 2017-05-24 礼进生物医药科技(上海)有限公司 PD-1 resisting antibody and treatment application thereof
WO2017117464A1 (en) 2015-12-30 2017-07-06 Kodiak Sciences Inc. Antibodies and conjugates thereof
CN109219618B (en) 2016-01-21 2022-08-09 辉瑞大药厂 Monospecific and bispecific antibodies against epidermal growth factor receptor variants III and CD3 and uses thereof
US20210309965A1 (en) 2016-03-21 2021-10-07 Dana-Farber Cancer Institute, Inc. T-cell exhaustion state-specific gene expression regulators and uses thereof
IL262404B2 (en) 2016-05-13 2024-04-01 Bioatla Llc Anti-ror2 antibodies, antibody fragments, their immunoconjugates and uses thereof
US11344619B2 (en) 2016-05-16 2022-05-31 Access To Advanced Health Institute Formulation containing TLR agonist and methods of use
MX2021010105A (en) 2016-06-01 2022-06-16 Infectious Disease Res Inst Nanoalum particles containing a sizing agent.
AU2017290805B2 (en) 2016-07-01 2023-11-16 Research Development Foundation Elimination of proliferating cells from stem cell-derived grafts
CA3034643A1 (en) 2016-09-20 2018-03-29 Ellen Weisberg Compositions and methods for identification, assessment, prevention, and treatment of aml using usp10 biomarkers and modulators
WO2018080573A1 (en) 2016-10-28 2018-05-03 Massachusetts Institute Of Technology Crispr/cas global regulator screening platform
WO2018148246A1 (en) 2017-02-07 2018-08-16 Massachusetts Institute Of Technology Methods and compositions for rna-guided genetic circuits
MX2019010458A (en) 2017-03-03 2020-01-20 Rinat Neuroscience Corp Anti-gitr antibodies and methods of use thereof.
JP7177076B2 (en) 2017-03-16 2022-11-22 ファイザー・インク tyrosine prototrophy
US11584790B2 (en) 2017-04-14 2023-02-21 Kodiak Sciences Inc. Complement factor D antagonist antibodies and conjugates thereof
MY201312A (en) 2017-06-02 2024-02-16 Pfizer Antibodies specific for flt3 and their uses
US11820822B2 (en) 2017-06-06 2023-11-21 Dana-Farber Cancer Institute, Inc. Methods for sensitizing cancer cells to T cell-mediated killing by modulating molecular pathways
GB2601923B8 (en) 2017-06-13 2023-03-08 Bostongene Corp Systems and methods for generating, visualizing and classifying molecular functional profiles
BR112020000679A2 (en) 2017-07-13 2020-07-14 Massachusetts Institute Of Technology targeting the hdac2-sp3 complex to enhance synaptic function
WO2019016784A1 (en) 2017-07-21 2019-01-24 Universidade De Coimbra Anti-nucleolin antibody
EP3684421A4 (en) 2017-09-18 2021-08-04 Children's Hospital Medical Center A strong insulator and uses thereof in gene delivery
CA3078460A1 (en) 2017-10-04 2019-04-11 Opko Pharmaceuticals, Llc Articles and methods directed to personalized therapy of cancer
PE20210708A1 (en) 2018-02-01 2021-04-16 Pfizer ANTIBODIES SPECIFIC TO CD70 AND THEIR USES
SG11202006883QA (en) 2018-02-01 2020-08-28 Pfizer Chimeric antigen receptors targeting cd70
CA3034912A1 (en) 2018-02-28 2019-08-28 Pfizer Inc. Il-15 variants and uses thereof
US12071476B2 (en) 2018-03-02 2024-08-27 Kodiak Sciences Inc. IL-6 antibodies and fusion constructs and conjugates thereof
US11525010B2 (en) 2018-05-23 2022-12-13 Pfizer Inc. Antibodies specific for GUCY2c and uses thereof
MX2020012539A (en) 2018-05-23 2021-02-16 Pfizer Antibodies specific for cd3 and uses thereof.
EP3817763A4 (en) 2018-07-03 2022-04-13 SOTIO Biotech Inc. Chimeric receptors in combination with trans metabolism molecules enhancing glucose import and therapeutic uses thereof
MX2021001771A (en) 2018-08-14 2021-07-16 Sotio Llc Chimeric antigen receptor polypeptides in combination with trans metabolism molecules modulating krebs cycle and therapeutic uses thereof.
BR112021003670A2 (en) 2018-08-28 2021-05-18 Vor Biopharma, Inc. genetically modified hematopoietic stem cells and their uses
US12084500B2 (en) 2019-01-23 2024-09-10 New York University Antibodies specific to delta 1 chain of T cell receptor
US20220289854A1 (en) 2019-04-30 2022-09-15 Dana-Farber Cancer Institute, Inc. Methods for treating cancer using combinations of anti-cx3cr1 and immune checkpoint blockade agents
JP2022538499A (en) 2019-07-03 2022-09-02 ボストンジーン コーポレイション Systems and methods for sample preparation, sample sequencing, and bias correction and quality control of sequencing data
CA3153932A1 (en) 2019-10-07 2021-04-15 Jonathan Kipnis Modulating lymphatic vessels in neurological disease
JP2022553640A (en) 2019-10-10 2022-12-26 コディアック サイエンシーズ インコーポレイテッド Methods of treating eye disorders
US20240092935A1 (en) 2019-10-11 2024-03-21 Beth Israel Deaconess Medical Center, Inc. Anti-tn antibodies and uses thereof
WO2021151079A1 (en) 2020-01-24 2021-07-29 University Of Virginia Patent Foundation Modulating lymphatic vessels in neurological disease
WO2021205325A1 (en) 2020-04-08 2021-10-14 Pfizer Inc. Anti-gucy2c antibodies and uses thereof
EP4146688A1 (en) 2020-05-06 2023-03-15 CRISPR Therapeutics AG Mask peptides and masked anti-ptk7 antibodies comprising such
JP2023533793A (en) 2020-07-17 2023-08-04 ファイザー・インク Therapeutic antibodies and their uses
WO2022023972A1 (en) 2020-07-30 2022-02-03 Pfizer Inc. Cells having gene duplications and uses thereof
EP4229222A2 (en) 2020-10-19 2023-08-23 Dana-Farber Cancer Institute, Inc. Germline biomarkers of clinical response and benefit to immune checkpoint inhibitor therapy
WO2022104104A2 (en) 2020-11-13 2022-05-19 Dana-Farber Cancer Institute, Inc. Personalized fusion cell vaccines
US20220180972A1 (en) 2020-12-04 2022-06-09 Bostongene Corporation Hierarchical machine learning techniques for identifying molecular categories from expression data
WO2022159793A2 (en) 2021-01-25 2022-07-28 Dana-Farber Cancer Institute, Inc. Methods and compositions for identifying neuroendocrine prostate cancer
US20220372580A1 (en) 2021-04-29 2022-11-24 Bostongene Corporation Machine learning techniques for estimating tumor cell expression in complex tumor tissue
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
WO2023012627A1 (en) 2021-08-02 2023-02-09 Pfizer Inc. Improved expression vectors and uses thereof
CA3232833A1 (en) 2021-09-27 2023-03-30 Kathleen Mcginness Chimeric receptor polypeptides in combination with trans metabolism molecules that re-direct glucose metabolites out of the glycolysis pathway and therapeutic uses thereof
CN118488969A (en) 2021-11-16 2024-08-13 舒迪安生物技术公司 Treatment of patients with myxoid/round cell liposarcoma
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
AU2023213817A1 (en) 2022-01-31 2024-08-01 Bostongene Corporation Machine learning techniques for cytometry
KR20240139082A (en) 2022-02-02 2024-09-20 화이자 인코포레이티드 Cysteine prototrophy
AU2023221961A1 (en) 2022-02-16 2024-09-26 Dana-Farber Cancer Institute, Inc. Methods for decreasing pathologic alpha-synuclein using agents that modulate fndc5 or biologically active fragments thereof
WO2024015561A1 (en) 2022-07-15 2024-01-18 Bostongene Corporation Techniques for detecting homologous recombination deficiency (hrd)
WO2024040208A1 (en) 2022-08-19 2024-02-22 Sotio Biotech Inc. Genetically engineered immune cells with chimeric receptor polypeptides in combination with multiple trans metabolism molecules and therapeutic uses thereof
WO2024040207A1 (en) 2022-08-19 2024-02-22 Sotio Biotech Inc. Genetically engineered natural killer (nk) cells with chimeric receptor polypeptides in combination with trans metabolism molecules and therapeutic uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991019798A1 (en) * 1990-06-20 1991-12-26 Dana Farber Cancer Institute Vectors containing hiv packaging sequences, packaging defective hiv vectors, and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991019798A1 (en) * 1990-06-20 1991-12-26 Dana Farber Cancer Institute Vectors containing hiv packaging sequences, packaging defective hiv vectors, and uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
See also references of WO9325698A1 *
VIROLOGY, vol. 152, no. 1, 1986, ORLANDO US, pages 268-271, XP002021491 BANAPOUR, B. ET AL.: "The Acquired Immunodeficiency Syndrome associated retrovirus is not sensitive to lysis or inactivation by human serum " *

Also Published As

Publication number Publication date
WO1993025698A1 (en) 1993-12-23
CA2137361A1 (en) 1993-12-23
JPH09507741A (en) 1997-08-12
EP0644946A4 (en) 1997-03-12

Similar Documents

Publication Publication Date Title
WO1993025698A1 (en) Vector particles resistant to inactivation by human serum
WO1993025698A9 (en) Vector particles resistant to inactivation by human serum
EP0746625B1 (en) Targetable vector particles
US5449614A (en) Recombinant retroviruses with amphotropic and ecotropic host ranges
US5925345A (en) Vectors including foreign genes and negative selective markers
Shimada et al. Targeted and highly efficient gene transfer into CD4+ cells by a recombinant human immunodeficiency virus retroviral vector.
Buchholz et al. In vivo selection of protease cleavage sites from retrovirus display libraries
Robbins et al. Increased probability of expression from modified retroviral vectors in embryonal stem cells and embryonal carcinoma cells
CA1315719C (en) Retroviral packaging cell lines and processes of using same
US5672510A (en) Retroviral vectors
Kim et al. Construction of retroviral vectors with improved safety, gene expression, and versatility
US5747323A (en) Retroviral vectors comprising a VL30-derived psi region
JP2003530064A (en) Coat protein modified baculovirus-vector for gene therapy
US5554524A (en) More complex type retroviruses having mixed type LTR, and uses thereof
US5576201A (en) Retroviral vector particles for transducing non-proliferating cells
CA2149889A1 (en) Cell-type specific gene transfer using retroviral vectors containing antibody-envelope fusion proteins
JP3908274B2 (en) Vector having a therapeutic gene encoding an antimicrobial peptide for gene therapy
US5580766A (en) Retroviral vector particles for transducing non-proliferating cells
US5736360A (en) Endothelial cell tropic compositions and methods of making and using the same
US5837503A (en) Vector containing viral gene transcribed by RNA polymerase III and methods for use
Wolff et al. Tissue tropism of a leukemogenic murine retrovirus is determined by sequences outside of the long terminal repeats.
EP0511311A1 (en) Novel retroviral vectors
US6762031B2 (en) Targeting viral vectors to specific cells
Antoine et al. Envelope and long terminal repeat sequences of an infectious murine leukemia virus from a human SCLC cell line: implications for gene transfer
Baum et al. Contribution of the retroviral leader to gene transfer and expression in packaging cells and myeloid stem and progenitor cells

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19941207

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

RHK1 Main classification (correction)

Ipc: C12N 15/86

A4 Supplementary search report drawn up and despatched
AK Designated contracting states

Kind code of ref document: A4

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LI LU MC NL PT SE

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 19971201